Plantas Medicinales Con Acetogeninas antiCA
Plantas Medicinales Con Acetogeninas antiCA
Xuefei Yang
Sreeja Puthanpura Sasidharan Editors
Bioprospecting
of Tropical
Medicinal
Plants
Bioprospecting of Tropical Medicinal Plants
Karuppusamy Arunachalam • Xuefei Yang
Sreeja Puthanpura Sasidharan
Editors
Bioprospecting of Tropical
Medicinal Plants
Editors
Karuppusamy Arunachalam Xuefei Yang
Center for Studies in Stem Cells Key Laboratory for Wild Plant Resources
Cell Therapy and Toxicological Kunming Institute of Botany
Genetics (CeTroGen) Kunming, Yunnan, China
Graduate Program in Health and
Development in the Midwest Region
Faculty of Medicine (FAMED)
Federal University of Mato
Grosso do Sul (UFMS)
Campo Grande, Cidade Universitária
Pioneiros, MS, Brazil
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland
AG 2023, Corrected Publication 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by similar
or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Switzerland AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
v
vi Preface
We also thank our colleagues and our research students for their cooperation and
critical suggestions. The technical support and continued encouragement received
from Dr. Kenneth Teng (Editor) and the book publishing team at Springer Nature,
Springer New York 233, Spring Street, New York, NY 10013-1522, USA, is also
acknowledged. We express deep gratitude to Kunming Institute of Botany (KIB),
Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Fundação
de Apoio ao Desenvolvimento do Ensino (FUNDECT), and Universidade Federal
de Mato Grosso do Sul (UFMS) for financial support. Also, this project work was
financially supported by the Chinese Academy of Sciences – President’s International
Fellowship for Postdoctoral Research (CAS-PIFI, Reference no. 2020PB0112),
Inventory and Database Construction Project of Herbal Medicine, along with the
“Belt and Road Countries” (Grantnumber 2018FY100700), the Southeast Asia
Biodiversity Research Institute, Chinese Academy of Sciences, Peoples Republic of
China (GrantnumberY4ZK111B01), and the Yunnan Province Science and
Technology Department (Grantnumber 202203AP140007). Finally, we acknowl-
edge Almighty God, who provided all the channels to work in cohesion and coordi-
nation right from the conception of the idea to the development of the final version
of this book.
vii
viii Contents
Appraisal of Medicinal Plants for Pharmacological Properties���������������� 1061
Mehmet Zeki Kocak and Mustafa Güven Kaysim
Pharmacological Properties and Tissue Culture Method
of Endangered Medicinal Plants������������������������������������������������������������������ 1081
Yuhong Zheng, Xin Shi, and Li Fu
Natural Compounds with Pharmacological Properties
in Clinical Trials �������������������������������������������������������������������������������������������� 1097
Morvarid Noormohammadi and Farzad Shidfar
Phytopharmacological Aspects of the Genus Terminalia���������������������������� 1117
Aswathi Pokkadath, Reshmi Chembrammal, and John Ernest Thoppil
Anticancer Potential of Plant-Derived Compounds:
An Overview of Their Epigenetic Mode of Action�������������������������������������� 1135
Priyanka Soni, Md. Sajid Ghufran, and Govinda Rao Duddukuri
Treatment of Cancer Using Combination of Herbal and Novel
Drug Delivery System������������������������������������������������������������������������������������ 1177
Nikita Kale
Anti-inflammatory Potential of Lead Compounds and Their
Derivatives from Medicinal Plants �������������������������������������������������������������� 1199
Nisha Sam Nirmala, Navina Bala Krishnan, Vaishnavi Vivekanandan,
and Krishnaraj Thirugnanasambantham
Alzheimer’s Disease Treatment Using Natural Foods:
A Overview ���������������������������������������������������������������������������������������������������� 1233
Tanima Bhattacharya, Debashrita Das, Hitesh Chopra,
and Atif Amin Baig
Natural Products Used in the Treatment of Autoimmune Disorder���������� 1247
Anjali Saharan, Meenakshi Dhanawat, Chander Parkash Dora,
Rakesh Kumar Sindhu, and Inderjeet Verma
Strategies to Improve Antimicrobial Activity of Natural Products:
Approaches and Challenges�������������������������������������������������������������������������� 1265
Cristina M. Pérez Zamora, Carola A. Torres, and Ana M. Gonzalez
Plants with Immunomodulatory Potential Described in Ayurveda���������� 1299
Sinimol Peethambaran Thekkekkoottumughath
Plant-Derived Drugs for Alzheimer’s Disease and Other
Neurological Disorders���������������������������������������������������������������������������������� 1327
B. Sumithra, Sanjeeb Kumar Mandal, Bishwambhar Mishra,
K. V. S. S. N. Mounika, J. Caleb Joel Raj, and C. V. S. Aishwarya
xii Contents
Properties and Mechanism of Antimicrobial Agents from
Plant-Derived Essential Oils ������������������������������������������������������������������������ 1347
Afroze Naveed Basha, Ramya Subramanian, Kandeepan Chithan,
Gopinath Gurulingam Vincent, Karthigeyan Murugesan,
Ananthavalli Ramachandran, Sivakumar Pethanan, Mani Panagal,
Chella Perumal Palanisamy, and Ramaraj Jayakumararaj
Nanotechnological Modus Operandi for the Delivery of Cytotoxic
Phytochemicals���������������������������������������������������������������������������������������������� 1365
Thomson Alex, Alankar Shrivastava, Damanpreet Kaur Lang,
Rakhi Khabiya, Sweta S. Koka, and Yasmin Sultana
Plant-Based Green Nanoparticles in Cancer Diagnosis and
Chemotherapy������������������������������������������������������������������������������������������������ 1387
Arun John and Rinu Elizabeth Roy
Use of Plant-Derived Nanoparticles in Cancer Therapy���������������������������� 1405
R. Sai Nandhini, S. Kalpana Shree, Phalguni Maity, G. S. Madhumathi,
Anindita Bhar, and Jeyanthi Palanivelu
The Global Concern for Cancer Emergence and Its Prevention:
A Systematic Unveiling of the Present Scenario������������������������������������������ 1429
Md. Sajid Ghufran, Priyanka Soni, and Govinda Rao Duddukuri
Commonly Used Poisonous Medicinal Plants in Unani System
of Medicine ���������������������������������������������������������������������������������������������������� 1457
Shaikh Ajij Ahmed Makbul and Sayeedur Rahman
Herbal Drugs: Safety, Cost-Effectiveness, Regulation,
Current Trends, and Future Directions ������������������������������������������������������ 1479
Sidra Nisar Ahmed, Mushtaq Ahmad, Mohammad Zafar, Ghulam Yaseen,
Nadia Iqbal, Neelum Rashid, Samina Kousar, and Adeela Haroon
Therapeutic Properties of Herbal Constituents Subjected for Clinical
Trials���������������������������������������������������������������������������������������������������������������� 1495
Esha Vatsa and Mehak Aggarwal
Plant-Derived Immunomodulators Targeting COVID-19
(SARS-CoV-2): Preclinical Evaluation and Clinical Trials������������������������ 1515
Robin, Pardeep Kaur, Jagdeep Kaur, Kamaljit Kaur, and Sunidhi Miglani
Correction to: Bioprospecting of Tropical Medicinal Plants �������������������� C1
Karuppusamy Arunachalam, Xuefei Yang,
and Sreeja Puthanpura Sasidharan
Index���������������������������������������������������������������������������������������������������������������� 1533
About the Authors
xiii
xiv About the Authors
xv
xvi Abbreviations
NO nitric oxide
NOS nitric oxide synthase
NSAIDs non-steroidal anti-inflammatory drugs
ONC oncology
OSCC squamous cell carcinoma
PGE2 prostaglandin E2
PGG2 prostaglandin G2
PGH2 prostaglandin H2
PNPs polymeric nanoparticles
PRC polycomb repressive complexes
RFC relative frequency of citation
RNS reactive nitrogen species
ROS reactive oxygen species
RSD respiratory systems diseases
SARS-CoV-2 severe acute respiratory syndrome coronavirus-2
SBDD structure-based drug design
SEM scanning electron microscopy
SMSD skeleto-muscular system disorders
SOD superoxide dismutase
SPE solid phase extraction
SRBC sheep red blood cells
STZ streptozotocin
SUZ12 suppressor of zeste 12 homologue
TBHQ tertiary butylhydroquinone
TEM transmission electron microscopy
TET ten eleven translocases
Th helper T cells
THC delta-9-tetrahydrocannabinol
TLC thin layer chromatography
TM traditional medicine
TNF-α tumor necrosis factor-α
TSGs tumor suppressor genes
UC ulcerative colitis
UV use value
WBC white blood cells
WHO World Health Organization
Ethnobotanical Study of Medicinal Herbs
Used by the Naga Tribes of Eastern
Himalayas
1 Introduction
K. Semy (*)
Department of Botany, Don Bosco College Kohima, Kohima, India
R. Kuotsu
Research Scholar, Department of Botany, Nagaland University, Lumami, Nagaland, India
available for the disposal of human needs. Furthermore, the extraction and develop-
ment of various medicines and chemotherapeutics from these plants, as well as from
traditionally used rural herbal remedies, has been linked to an increased reliance on
medicinal plants in industrialized countries.
Northeast Indian states are some of the richest repositories of medicinal and
aromatic plants in the world [6]. This region has high medicinal plant diversity due
to variance in topography and physiognomy [7]. Nagaland is a part of the Indo-
Burma mega biodiversity hotspot, which includes an immense variety of plant spe-
cies and is one of the wealthiest in terms of biological wealth and endemism in the
Indian subcontinent [8]. Naga’s are part of the mongoloid race and since time
immemorial, forest products have aided in the socio-economic life of the Naga
tribes in their ever-growing demands for medicines, food and shelter. They have an
inextricable link with their forest and regard it as a provider, guide, healer and pro-
tector like most of the North-eastern tribal communities of India. Naga villages
encompass parts of the hilly Eastern Himalayas and are secluded from other cities,
and hence the local inhabitants developed a sense to rely on indigenous healing
knowledge using medicinal plants. However, tribal’s knowledge on therapeutic
herbs of northeast India and in particular Nagaland, when compared to the rest of
the country is still understudied. Meanwhile, the traditional knowledge is rapidly
degrading with the advancement of technology and modernization. During the ear-
lier years, many researchers have contributed to the studies on medicinal plants
from in and around Northeast India, Assam [9–11], Manipur [12, 13], Arunachal
Pradesh [14–17] and Nagaland [18–24]. However, in contrast to those documented
plants focused on the three plant forms (herb, shrub and trees), the present investiga-
tion has been chiefly focused on the medicinal herbs prevalent in Nagaland. In con-
cern with the growing awareness and need for medicinal herbs, documentation of
plants is of vital importance. As a result, the current study was conducted in the
region with the following aims: (1) To document medicinal uses of plants, their rela-
tive importance and information for future investigation in novel drug applications
and (2) to educate locals about the area’s declining wealth on traditional and medici-
nal flora.
2.1 Study Area
Nagaland lies in the North-eastern part of India between the latitude of 250 06´ N
and 270 04´ N and longitude of 930 20′ E and 950 15′ E. The state covers a geo-
graphical area of 16,579 km2 and is bordered by Assam in the northwest, Myanmar
and Arunachal Pradesh in the east and Manipur in the south. The state experience a
sub-tropical to warm temperate monsoonal climate with four seasons, viz., winter,
spring, summer and autumn. Annually, rainfall ranges between 1800 and 2500 mm
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas 3
with July and August receiving the highest rainfall. Temperature may rise from 21
to 36 °C in summer and drops from 21 to 4 °C during winter months. Frost is com-
mon at high elevations with snowfall in certain places and strong northwest wind
blows across the state with the onset of spring season. About 70% of the state’s
economy is dependent on agriculture along with other prominent economic activi-
ties including forestry, tourism and miscellaneous cottage industries. The recorded
forest cover of the state in 2021 is 8629.30 km2 which is about 52% of its geographi-
cal area. Tropical and sub-tropical evergreen forests accounts to one-sixth of the
forest area including palms, bamboo and timber and mahogany forests.
The history of medicinal plants is as long as the history of humans. Nagas have been
practicing the use of medicinal plants since time immemorial which has been passed
down from generation to generation through traditional practices and oral lore.
Before Christianity, Nagas practiced Totemism and Animism, which are primarily
indulged with worshipping nature. The core principle of this devotion is abided in
respecting and preserving nature through rituals and sustainable norms laid down
by the village priest. Supplementing such traditions, the practices of therapeutic
healing is also associated with the principles and beliefs of the tribes. In the present
study, a total of 161 herbaceous plant species belonging to 58 families and 127 gen-
era were documented during the survey. All the recorded herbs have certain medici-
nal values and have been extensively used by the tribes for treating several diseases
and ailments. The collected plant samples arranged in alphabetical order along with
their botanical and vernacular names, plant parts used, preparation and purpose/
mode of administration are enumerated in Table 1. The family Asteraceae (24) was
reported with the largest number of species followed by Lamiaceae (11), Poaeaea
(9), Solanaceae (7), Leguminosae, Polygonaceae, Malvaceae and Rubiaceae (6
4
(continued)
5
6
Table 1 (continued)
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
20. Bidens pilosa L. Zorha Asteraceae Leaves Fresh paste applied on Blood coagulation, wounds, burns,
wounds; dried leaves taken malaria, and arthritis.
orally
21. Breynia retusa (Dennst.) – Phyllanthaceae Leaves and Consumed either fresh or Diabetes, antioxidant, reduce
Alston roots dried blood sugar.
22. Brugmansia suaveolens Lalho Solanaceae Leaves Leaves wrapped in banana Ulcers, tonsillitis, body ache.
(Humb. & Bonpl. ex leaf warmed in fire and
Willd.) Bercht. & J.Presl dapped on affected area to
ease pain
23. Cannabis sativa L. Ganja Cannabaceae Leaves Leaf paste used as hemostatic Cuts and wounds, malaria,
to cuts and wounds; decoction stomach ache.
taken orally
24. Cardamine hirsuta L. Seguoga Brassicaceae Whole plant Cooked with other herbs and Digestive disorder.
eaten with rice
25. Catharanthus roseus (L.) Tsuinrinaro Apocyanaceae Leaves Leaf extract applied on Skin disease, irregular blood
G.Don infected skin; boiled and pressure, stomach problems.
consumed in small quantity
with Neem
26. Centella asiatica (L.) Urb. Gara Apiaceae Whole plant Boiled or raw leaves Diabetic, wounds, burns, known
Fresh to improve memory power,
diarrhoea, constipation.
27. Chloris barbata Sw. – Poaceae Leaves Leaf paste applied on infected Skin infection, antimicrobial.
area
28. Chromolaena odorata (L.) – Asteraceae Leaves Leaf paste or juice rubbed on Fresh wounds, burns, skin
R.M.King & H.Rob. wounds infection.
29. Clerodendrum bracteatum Khriehenyii Lamiaceae Leaves Juice is applied on scalp; paste Astringent, remove dandruff.
Wall. ex Walp. taken orally
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
30. Clerodendrum Gathere Lamiaceae Leaves Boiled leaves are eaten Lowers blood pressure.
glandulosum Lindl.
31. Clerodendrum serratum Atsuksuba Lamiaceae Leaves Decoction of leaves are Irregular menstruation.
(L.) Moon consumed
32. Clerodendrum villosum Akawa Lamiaceae Leaves Decoction is drunk; juice Kill lice, liver problems.
Blume massage on scalp
33. Coix lacryma-jobi L. Kesi Poaceae Fruit Fruits soaked in water Warts, appendicitis, lungs
overnight and taken orally disorder, arthritis.
34. Commelina benghalensis Akhovepii Commelinaceae Aerial parts Fresh leaves and roots used as Sun burns, anti- inflammatory,
L. poultice diuretic, laxative.
35. Colocasia esculenta (L.) Dziinuo Araceae Leaves and Cooked and consumed Anti-inflammatory, anti-fungal,
Schott tubers inhibit tumor and gastrointestinal
problems.
36. Costus speciosus Thevobuoto Costaceae Stem and Boiled or soaked rhizome in Skin infection, anti- cancer,
(J.Koenig) Sm. rhizome water and consumed; stem anti-microbial, obesity, urinary
paste applied on infested skin tract infection.
with maggots
37. Crassocephalum – Asteraceae Leaves Extract used orally with honey Indigestion, oral problems,
crepidioides (Benth.) S. epilepsy, inflammation.
Moore
38. Crotalaria juncea L. – Leguminosae Root, pod and Concoction of root, pods and Skin diseases, rashes, high blood
leaves leaves applied externally or pressure, and astringent.
ingested in small dosage
39. Curculigo capitulata Koritong Hypoxidaceae Root, flowers Decoction of young plant Conjunctivitis, ear-ache,
(Lour.) Kuntze and leaves parts; root soaked overnight in antiseptic, haemostatic.
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
Table 1 (continued)
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
40. Curculigo orchioides Koritong Hypoxidaceae Root, flowers Decoction of young plants Ear-ache, antiseptic.
Gaertn. and leaves with lime
41. Curcuma angustifolia Chiecie Zingiberaceae Flower and Consumed both fresh and Fever, cough, antifungal,
Roxb. rhizome dried with other herbs analgesics, anticancer.
42. Curcuma aromatica Salisb. Hutou Zingiberaceae Flower and Taken both fresh and dried Gastrointestinal disorder, arthritis,
rhizome with therapeutic herbs skin infections and rashes.
43. Cuscuta chinensis Lam. Tsuiali Convolvulaceae Aerial parts Paste applied on pain affected Anti-depression, urinary bladder
area; juice taken orally problem, liver ailments, joint pain.
44. Cuscuta reflexa Roxb. Tsuiali Convolvulaceae Aerial parts Juice extract is boiled and Urination disorder, muscle cramp,
consumed; paste applied on cold cough, warts, carminative.
affected area
45. Cyanotis vaga (Lour.) – Commelinaceae Whole plant Plant extract is ingested in Liver dysfunction, weight loss,
Schult. & Schult.f. small quantity with honey enhance energy metabolism.
46. Cyperus iria L. – Cyperaceae Leaves Boiled with honey and juice is Stomach ache, diuretic, regulate
consumed menstruation, cure itching.
47. Datura innoxia Mill. Lhalho Solanaceae Flower and Either fresh or dried use as Malaria, antiseptic, cardiac arrest,
leaves poultice stomach ulcers.
48. Dicranopteris linearis Kajangtong Gleicheniaceae Leaves Poultice on painful body parts Fever, wounds, asthma, sores,
(Burm.f.) Underw. ulcers.
49. Dioscorea alata L. Thecu Dioscoreaceae Tuber Boiled and taken Gastritis, constipation, stomach
ache.
50. Diplazium esculentum Gasiilo Athyriaceae Leaves Taken either boiled or raw Measles, gastrointestinal, diabetes,
(Retz.) Sw. with aromatic rice and pond glandular swelling, bone fracture,
salt constipation.
51. Drymaria cordata (L.) Pfiipfiinyii Caryophyllaceae Whole plant Squeezed along with mustard Sinusitis, migraine, bronchitis.
Willd. ex Schult. oil and juice applied on
infected area; taken orally
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
52. Eclipta prostrata (L.) L. Bhringraj Asteraceae Leaves Extract mixed with honey, Hepatitis, snake bites, jaundice,
milk and ingested; leaf paste liver tonic, asthma, respiratory
rubbed on wounds ailments.
53. Elatostema sessile Gajo Urticaceae Leaves Leaves boiled and eaten with Gastrointestinal problems.
J.R.Forst. & G.Forst. rice
54. Eleusine indica (L.) – Poaceae Whole plant Boiled and drunk Laxative, hypertension, fever, cold
Gaertn. cough, malaria, relieves pain,
astringent, asthma.
55. Elsholtzia blanda (Benth.) Niepfii Lamiaceae Leaves and Consumed both fresh and Reduce tonsil swell, fever, cough,
Benth. flower dried and maintain blood pressure.
56. Entada pursaetha DC. Tholi Leguminosae Seed Cleansed in running water for Burns, jaundice in children,
several days, roasted and after-effects of stroke, helps in
consumed in small dosage. blood circulation, parasitic
infection.
57. Equisetum arvense L. Siihie Equisetaceae Aerial parts Boiled with water and lemon Kidney stone, bladder problems,
juice tuberculosis, heal ulcer.
58. Erigeron linifolius Willd. – Asteraceae Aerial parts Plant paste applied on infected Antibacterial, anti-inflammatory.
area
59. Eryngium foetidum L. Dunia Apiaceae Whole plant Consume both fresh and Infertility complications, ear ache,
cooked with other herbs de-worming, malaria.
60. Eupatorium adenophorum Japan nha Asteraceae Aerial parts Fresh paste applied on Anti-inflammatory, anti-microbial,
Spreng. infected area analgesic, blood coagulation.
61. Euphorbia heterophylla L. – Euphorbiaceae Leaves Boiled with pond salt and Asthma, anti-cancer, diarrhea.
consumed
62. Euphorbia hirta L. Dudiya Euphorbiaceae Aerial shoots Consumed either fresh or Cold cough, bronchitis,
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
63. Fagopyrum esculentum Garei Polygonaceae Aerial parts Either fresh or dried young Cardiovascular, antioxidant,
Moench leaves cooked with local rice stomach congestion, diabetic.
and consumed
64. Galinsoga parviflora Cav. Bomara Asteraceae Leaves Fresh paste applied on Eczema, skin infection, rashes,
infected area bleeding wounds.
65. Gomphrena celosioides – Amaranthaceae Whole plant Extract taken orally; fresh Gastrointestinal, breathlessness,
Mart. paste applied on skin rashes skin rashes.
66. Gynura bicolor (Roxb. ex Liezienuo Asteraceae Aerial parts Young leaves boiled with local Constipation, diabetes, post-
Willd.) DC. rice and taken orally labour recovery, migraines,
haemoptysis.
67. Hibiscus sabdariffa L. Gakhro Malvaceae Leaves and Consumed either dried-fresh Blood pressure, aids in digestion,
flower or made into porridge with skin rashes, inflammatory, food
rice poisoning.
68. Hibiscus syriacus L. Chakha ga Malvaceae Flower and Flower paste applied on scalp; Antifungal, hair and skin
leaves young leaves boiled and eaten treatment, gastrointestinal.
69. Hodgsonia macrocarpa Ketsamo Cucurbitaceae Seeds Roasted seeds mixed with Bacterial infection in feet, heal
(Blume) Cogn. aromatic herbs, pond salt and wounds, fever, nose ulcers, cure
consumed burns.
70. Houttuynia cordata Thunb. Gatha Saururaceae Whole plant Fresh plant mixed with Diuretic action, detoxification,
fermented soya bean and hypertension, improve appetite.
eaten
71. Hydrocotyle javanica – Araliaceae Leaves and Crushed leaf paste applied on Gastritis, eye infection, fresh cut
Thunb. tender shoots wounds: young shoots cooked wounds.
and consumed
72. Impatiens latiflora Hook.f. Ciikanyii Balsaminaceae Leaves Fresh paste is applied on skin Insect bites, allergies, indigestion,
& Thomson infection; leaves taken orally analgesic.
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
73. Imperata cylindrica (L.) Azu Poaceae Aerial shoots Paste is applied on wounds; Diuretic, gastrointestinal, urinary
Raeusch. leaves are consumed tract infection.
74. Ipomoea involucrata – Convolvulaceae Roots and Extract is taken orally Hypertension, diabetes, analgesic,
P. Beauv. leaves psychotomimetic, antimicrobial,
fatigue.
75. Ipomoea purpurea (L.) – Convolvulaceae Leaves and Plant extract taken orally Constipation, oedema in lungs,
Roth flower mental disorders, jaundice.
76. Ipomoea quamoclit L. Tamlata Convolvulaceae Leaves and Leaf paste applied on infected Insect bites, rashes, hairfall,
flower skin; young parts are antidiabetic, antimicrobial.
consumed
77. Justicia adhatoda L. Tsiesenyii Acanthaceae Leaves Decoction of young shoots Carminative, paralysis,
with therapeutic herbs gonorrhoea, chronic rheumatism.
78. Kaempferia rotunda L. Bhuichampa Zingiberaceae Flower and Paste applied on wounds; Stomach ache, food poisoning,
rhizome taken orally emetic, thermogenic.
79. Kalanchoe pinnata (Lam.) Tsatsovo Crassulaceae Leaves Fresh grinded with therapeutic Pulmonary infection, kidney
Pers. herbs and ingested stones, stomach ache, gastric
ulcer, allergies, diabetes.
80. Lantana camara L. Anitong Verbenaceae Leaves and Boiled with honey and juice Fever, cold cough, antimicrobial,
flowers taken in small dosage chicken pox, ulcers, skin rashes,
wheezing cough.
81. Leucas aspera (Willd.) Chota halkusa Lamiaceae Leaves Paste is applied on infected Antimicrobial, snake bites, insect
Link area bites.
82. Ludwigia perennis L. – Onagraceae Flower Fresh paste applied on Toothache and fever.
infected tooth
83. Lycopodium cernuum L. Mangrang Lycopodiaceae Aerial parts Dried-grinded, boiled with Constipation, chronic lung
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
84. Mentha arvensis L. Pudina Lamiaceae Aerial parts Consumed either fresh or Hypertension, heart disease,
boiled inflamed joints and arthritis.
85. Mikania micrantha Kunth Japanza Asteraceae Leaves Fresh leaves are grinded and Lower blood pressure; reduce
extract applied for sinus; blood sugar level, sinusitis.
extract taken orally
86. Mimosa pudica L. Keriinganha Leguminosae Root and leaves Freshly grinded and taken Asthma, jaundice, ulcer, small
orally pox, conjunctivitis.
87. Mirabilis jalapa L. Jamtangnaro Nyctaginaceae Roots and Concoction of roots and Purgative, cathartic inflammation,
leaves leaves taken orally antiviral, diuretic.
88. Mussaenda macrophylla Seirhobie Rubiaceae Fruits Boiled and juice consumed Antiviral, male impotency,
Wall. gastrointestinal problems
89. Ocimum sanctum L. Nieco Lamiaceae Aerial parts Mixed with ginger, honey and Asthma, eye sore, dysentery,
taken orally arthritis, gastritis, antimicrobial.
90. Ocimum tenuiflorum L. Nieco Lamiaceae Flower and Either dried, fresh and mixed Anti-ageing, cough, headache,
leaves with other herbs and acne, anticancer, heartache, fever,
consumed eye health.
91. Oenanthe javanica Gakra Apiaceae Young aerial Boiled or raw with soyabeans, Jaundice, abdominal pain,
(Blume) DC. parts garlic, ginger, pond salt and leukaemia, hepatitis.
eaten with aromatic red rice
92. Oxalis corniculata L. Keve Oxalidaceae Whole plant Fresh plants are eaten wholly Aids in digestion, antiseptic, fresh
cut wounds, burns.
93. Oxalis debilis Kunth Thezutsiituo Oxalidaceae Whole plant Fresh plants are eaten wholly Indigestion, constipation,
acid-reflux.
94. Paederia foetida L. Menyiero Rubiaceae Aerial parts Boiled with water, herbs, pond Gastrointestinal, abdominal pain,
salt and ingested rheumatism, stomach oedema,
gastritis, ulcers.
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
95. Panax ginseng C.A.Mey. Tsudiemozu Araliaceae Root, leaves Extract mixed with sorghum Heart problems, diabetes, cancer,
oil, pond salt, water and tuberculosis, ulcers.
drunk; dried powdered mixed
with honey and water
96. Panax pseudoginseng Wall. Takumtsu Araliaceae Root and leaves Dried or fresh root and leaves Anticancer, tuberculosis, diabetes.
mozu grounded and taken orally
97. Paris polyphylla Sm. – Melanthiaceae Rhizome Fresh or dried rhizome Diarrhoea, analgesic, burn, cut,
applied on infected area or anticancer, snake, spider and
taken orally scorpion bite, antispasmodic.
98. Paspalum distichum L. – Poaceae Whole plant Either fresh or dried plants are Bronchitis, arthritis, blood tonic,
boiled and ingested antibacterial.
99. Passiflora edulis Sims Bel Passifloraceae Leaves and Consumed either steamed or Asthma, snakebite, liver- tonic,
fruits fried; fruits eaten fresh or heart problems, maintains blood
fermented pressure, malaria.
100. Peperomia pellucida (L.) – Piperaceae Leaves Paste applied on infected skin; Anti-inflammatory, rheumatism,
Kunth eaten either fresh or boiled fatique, acne, boils.
101. Perilla frutescens (L.) Kenyie Lamiaceae Leaves and Leaves consumed either fresh Stomach problems, gastritis,
Britton seeds or steamed; seeds are roasted haemostatic.
made into paste and ingested
102. Persicaria chinensis (L.) Priizie Polygonaceae Whole plant Young leaves and stem are Antibacterial, snake bite, eye
H. Gross boiled with pond salt and treatment, mosquito repellent.
ingested
103. Persicaria hydropiper (L.) Priizie Polygonaceae Leaves Fresh leaves boiled and taken Neuro-protective effect, anti-
Delarbre cancer, against rheumatism.
104. Phyllanthus fraternus – Phyllanthaceae Whole plant Paste directly applied on Diuretic, laxative, gonorrhoea,
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
105. Physalis peruviana L. Chahacasi Solanaceae Fruit and leaves. Fresh fruits and leaves Antihistamine, antiviral, cancer,
grounded and taken in small asthma, dermatitis, gout, urinary
dosage tract disorder.
106. Pilea microphylla (L.) – Urticaceae Aerial parts Plant extract rubbed on Antibacterial, skin infection,
Liebm. infected skin antifungal, allergies.
107. Piper betle L. Seipanyii Piperaceae Leaves Fresh paste applied on wound Haemostatic.
108. Piper longum L. Pipali Piperaceae Leaves Eaten raw or steamed with Chronic malaria, spleen, tumours,
pond salt tongue paralysis, respiratory
infection.
109. Plantago asiatica L. Gapa Plantaginaceae Leaves Leaves are boiled and eaten Urinary tract infection, promote
urination, relieve phlegm
discomfort.
110. Plantago erosa Wall. Isabgol Plantaginaceae Whole plant Boiled with other herbs with Bleeding and inflammation,
local pond salt and taken with constipation, antibacterial,
aromatic rice indigestion.
111. Plantago major L. Gapa Plantaginaceae Whole plant Boiled and eaten Cuts and burns, sprains.
112. Polygonum molle D. Don Gazie Polygonaceae Leaves and Fresh leaves poultice on Anti-inflammatory, anticancer,
young stem infected area; young leaves skin rashes, astringent properties.
and stem taken orally
113. Polygonum plebeium R.Br. – Polygonaceae Leaves Taken either fresh or boiled Bowel movement, constipation,
stomach ailments.
114. Pouzolzia hirta Blume ex – Urticaceae Leaves Extracts are drunk Bowel movement, constipation,
Hassk. stomach ailments.
115. Pouzolzia zeylanica (L.) – Urticaceae Aerial parts Poultice on infected area Ulcers, syphilis, gonorrhoea,
Benn. galactagogue, stomachache.
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
116. Psophocarpus Charkona Leguminosae Fruit Consumed fresh mixed with Blood purifier, anti-oxidant,
tetragonolobus (L.) DC. pond salt, king chili and diabetes.
soybean
117. Pteridium aquilinum (L.) – Dennstaedtiaceae Roots Steam bath Diuretic, arthritis, old wound,
Kuhn aphrodisiac, de-worming.
118. Ricinus communis L. Louca Euphorbiaceae Leaves Dried leaves grounded and Backache, cramp, menstrual
taken in small dosage problems, constipation.
119. Rubia cordifolia L. Chenhu Rubiaceae Whole plant Extract or paste applied on Ringworm, leucoderma, skin
infected skin rashes disease.
120. Rubus ellipticus Sm. Ruomvii Rosaceae Berries and Grinded with water and Cold cough, sore throat,
leaves consumed indigestion, constipation, diuretic.
121. Rubus niveus Thunb. Temeirom Rosaceae Berries and Grinded with water and Snake bite, rheumatic,
leaves consumed detoxification, dysentery,
menstrual bleeding.
122. Rumex patientia L. Meza gakrie Polygonaceae Roots and Poultice on infected area; Constipation, fresh cut wounds,
leaves juice of roots and leaves taken skin rashes, aids in digestion.
in small dosage
123. Rungia pectinata (L.) Nees – Acanthaceae Leaves Fresh leaf paste applied on the Small pox, body ache, aperients
body effects.
124. Scutellaria rivularis Wall. Angamejep Lamiaceae Whole plant Paste mixed with sorghum oil Insect and spider bites.
ex Benth. talula and rubbed
125. Selaginella involvens (Sw.) – Selaginellaceae Aerial parts Taken orally with other herbs Internal haemorrhoid bleeding,
Spring blood expediting.
126. Senecio cappa Buch.-Ham. Mesakraza Asteraceae Whole plant Extract applied externally Skin disease, boils.
ex D.Don
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
(continued)
15
Table 1 (continued)
16
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
127. Senna hirsuta (L.) – Leguminosae Leaves Dried, grinded-applied on Dysentery, ringworm, skin
H.S.Irwin & Barneby infected skin and taken orally infection, germicide, antiparasite.
in small dosage
128. Sesamum orientale L. Pingnak Pedaliaceae Seeds Roasted, consumed, or applied Laxative, skin hydration, hair
on skin and scalp growth.
129. Setaria glauca (L.) P. – Poaceae Whole plant Boiled and ingested Fever, cough.
Beauv.
130. Setaria pumila (Poir.) – Poaceae Aerial parts Boiled with other herbs, Rheumatic, fever.
Roem. & Schult. mixed with salt and consumed
131. Sida acuta Burm. f. Bala Malvaceae Leaves Dried, grinded with oil and Asthma, tuberculosis, oral
taken orally problems, urinary tract infection,
testicular swelling.
132. Smilax zeylanica L. Fiipro Smilacaceae Leaves and Fresh paste rubbed on infected Skin infections; maintain blood
roots skin; boiled with cow milk pressure, indigestion, joint pain,
and ingested sexual vigor.
133. Solanum gilo Raddi Ciepfi Solanaceae Fruits Roasted in hot ash and eaten Aids in digestion, maintains blood
pressure.
134. Solanum nigrum L. Gadzii Solanaceae Fruits Consumed either fresh or Mouth ulcers, skin infections,
boiled with pond salt asthma, tonic, and cough.
135. Solanum torvum Sw. Tsociepfii Solanaceae Fruits Roasted in ash, fried with Cardiac arrest, ulcers, fever,
sorghum or boiled with pond cough, wounds, liver disorder.
salt
136. Solanum viarum Dunal Chiikrii Solanaceae Fruits Dried and mixed with other Anticancer,
therapeutic herbs and taken antifungal, anti- inflammatory.
orally
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
137. Sonchus arvensis L. Nhana Asteraceae Leaves, stem, Fresh leaves steamed and Cough, digestive disorder,
young roots eaten increase appetite and improve
eyesight.
138. Sonchus asper (L.) Hill Nhana Asteraceae Leaves and Young leaves cooked and Sedative, anti-cancer, blood
stems eaten purifier, antidepressant.
139. Spermacoce hispida L. – Rubiaceaea Aerial parts Consumed fresh young shoots Gallstones, conjunctivitis,
haemorrhoids, tonic.
140. Spermacoce keyensis Small – Rubiaceaea Aerial parts Young shoots consumed either Malaria, boils, haemorrhage, skin
fresh or dried infection.
141. Spermacoce latifolia Aubl. – Rubiaceae Aerial parts Dried and mixed with water Leukemia, digestive problems,
and taken orally skin rashes, urinary tract infection,
respiratory ailments.
142. Sphaeranthus indicus L. – Asteraceae Aerial parts Either dried or fresh mixed Skin infection, cough, diabetic,
with other herbs epilepsy.
143. Spilanthes acmella (L.) L. Kevenha Asteraceae Flower and Fresh flowers paste is applied Toothache, inflammation,
leaves on infected tooth.; leaves are pain-reliever, diuretic, gastric
boiled and taken orally ulcer.
144. Sporobolus diandrus – Poaceae Aerial parts Boiled and consumed orally Gonorrhoea, pain reliever, blood
(Retz.) P.Beauv. circulation.
145. Synedrella nodiflora (L.) – Asteaceae Leaves Dried, grinded and taken Cardiac problems, epilepsy, liver
Gaertn. orally disease, inflammatory.
146. Thunbergia coccinea Wall. Nulidongmoli Acanthaceae Tubers Extracts taken orally in small Aphrodisiac and tonic.
dosage
147. Thysanolaena latifolia Phipfe Poaceae Leaves and Plant extract mixed with lime, Treat boils, sore in eye, fever, and
(Roxb. ex Hornem.) Honda roots pond salt and ingested reduce inflammation.
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
(continued)
17
Table 1 (continued)
18
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
148. Tagetes erecta L. Puja niepou Asteraceae Leaves and Decoction of leaves Intestinal problems, gastritis,
flower mumps, sore eyes.
149. Taraxacum officinale (L.) – Asteraceae Whole plant Decoction and brewed for tea Reduce cholesterol, maintain
Weber ex F.H.Wigg. blood sugar level, improve
eyesight, cleans intestinal tract.
150. Tinospora sinensis (Lour.) Tsula Menispermaceae Stem Poulticed on affected area Applied to burns, bone fractures,
Merr. mesukzu sprains, stomach problems.
151. Tithonia diversifolia Mochitsiinara Asteraceae Leaves Paste applied on skin; dried or Antiseptic, indigestion, stomach
(Hemsl.) A.Gray fresh mixed with therapeutic ache, constipation, diabetes,
herbs ingested in small dosage menstrual pain.
152. Tridax procumbens (L.) L. – Asteraceae Leaves Paste rubbed on infected skin Anti-fungal, skin disease, insect
bites, anti-coagulant.
153. Triumfetta pilosa Roth Kuchii Malvaceae Leaves Dried and taken orally Inflammation, stomach ache,
jaundice.
154. Triumfetta rhomboidea Kuchii Malvaceae Leaves Dried and taken orally Gum problems, stomach ache,
Jacq. hepatitis, asthma.
155. Typha angustata Bory & – Typhaceae Shoot Concoction of leaves, honey, Male impotency, menstrual pain,
Chaub. ginger and consumed orally haemostatic.
156. Urena lobata L. Kouchii Malvaceae Root, stem and Concoction of leaves, root, Anticancer, anti-diabetic,
leaves stem with water and taken diarrhoea.
orally
157. Urtica dioica L. Zozie Urticaceae Leaves Leaves paste mixed with Hair fall, inflammations, prostate
essential oil and applied on cancer, hay fever, lower blood
scalp; leaves boiled and taken pressure, maintains blood sugar
orally level.
K. Semy and R. Kuotsu
Sl Vernacular
no. Scientific name name Family Parts used Preparation Purpose /usage/treatment
158. Wedelia chinensis Onnii Asteraceae Leaves Extract of leaves is rubbed on Hair growth, analgesic,
(Osbeck) Merr. scalp and ingested in minute antimicrobial, headache.
dosage
159. Zanthoxylum Ganyasei Rutaceae Leaves and Consumed either raw or Fever and toothache.
acanthopodium DC. fruits cooked with salt or soya
beans; paste applied on
infected tooth
160. Zanthoxylum oxyphyllum Ganyanyii Rutaceae Fruits, leaves, Poultice on infected area; Liver-tonic, toothache and fever.
Edgew. inflorescence cooked with sorghum oil,
pond salt and eaten with
aromatic rice
161. Zanthoxylum rhetsa DC. Ganya Rutaceae Fruits and Poultice on forehead; boiled Fever.
leaves and eaten with sticky rice
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas
19
20 K. Semy and R. Kuotsu
Most of the plants are processed in different ways like grinding, macerating, paste,
juice extracts and decoction in combination with other herbs or ingredients like
sorghum oil, pond salt, honey, milk and aromatic local rice. A warm poultice of
various medicinal herbs with sorghum oil is also applied to get relief from muscle
cramp, soreness, joint pain, body inflammation, body ache, fever and stomach con-
gestion. The gastronomic culture of Nagas involves in the intake of spicy and fer-
mented foods (bamboo shoots, soyabeans, mustard leaves, pickles, king chilies,
etc.) including smoked meat (pork, beef, mutton and wild animals). These habits
could have been a key reason for various health issues such as stomach pain, intes-
tinal disorders, asthma, chronic liver disease and lungs infection prevalent within
the tribal population. However, with these problems, the Nagas have developed a
sense to find sources in treating the associated ailments and thus majority of the
plants documented are known to be utilized for treating such gastrointestinal and
other associated ailments. In total, 34 herbs were recorded for treating gastrointes-
tinal problems (Acorus calamus, Alpinia malaccensis, Amaranthus spinosus,
Asparagus officinalis, Cannabis sativa, Catharanthus roseus, Curcuma aromatic,
Colocasia esculenta, Cyperus iria, Datura innoxia, Dicranopteris linearis,
Dioscorea alata, Diplazium esculentum, Elatostema sessile, Fagopyrum esculen-
tum, Gomphrena celosioides, Gynura bicolor, Hibiscus syriacus, Hydrocotyle
javanica, Kaempferia rotunda, Kalanchoe pinnata, Ocimum sanctum, Imperata
cylindrica, Mussaenda macrophylla, Paederia foetida, Polygonum plebium,
Pouzolzia hirta, P. zeylanica, Tagetes erecta, Tinospora sinensis, Tithonia diversifo-
lia, Triumfetta pilosa, T. rhomboidea and Typha angusta). From the present study, it
is evident that medicinal plants play a vital role in aiding to the health of indigenous
people in this region. Many of the tribes in the studied area still depend on the
medicinal plants for their daily healthcare routine over the modern system of medi-
cines. But over the years, this system may deteriorate due to the absence of interest
among the younger generations as most are influenced by modern technology and
their cultural importance are put aside. Moreover, the traditional healing systems
are passed down orally and hence detailed information is lost in the process.
Therefore, necessary steps should be taken up to encourage the upcoming genera-
tions as well as penned down the necessities in proper text for future references.
With the help of the government or village councils, robust regulations and manage-
ment plans should be laid to protect and educate the tribes about the benefits, medic-
inal purposes and conservation of these valuable herbs.
4 Conclusion
The study elucidated a very high number of therapeutic herbs accounting for 161
plants, which is one of the highest recorded data from Northeast India. According to
the current research, medicinal herbs will continue to play an essential role as a
health aid to the tribal communities of Eastern Himalayas. Traditional healing treat-
ments employing medicinal plants are widely used on a normative basis by the
22 K. Semy and R. Kuotsu
Nagas. Furthermore, due to the rising costs of personal health maintenance, herbal
therapies have grown more popular in these regions for treating various disorders.
However, as a result of developmental activities, deforestation, population growth
and indiscriminate exploitation, native medicinal plants are rapidly depleting and
have even lead to some plant species being endangered. Therefore, efforts must be
geared towards preserving them through sustainable management and enhance the
effectiveness, efficacy and rational use of medicinal plants, especially through the
integration into national, regional and local health policies and programmes. In situ
conservation techniques in home gardens and on-farm cultivation should be encour-
aged for socio-economic and sustainable growth. Also, these plants can fill the void
in the medical sector and become a new trend for upcoming research.
Acknowledgement The authors are thankful to the native healers, village elders, herbalist and
local inhabitants of Nagaland for sharing their traditional knowledge.
References
1. Davidson-Hunt I (2000) Ecological ethnobotany: stumbling toward new practices and para-
digms. Mod Assis Stat App 16:1–13
2. Mahesh B, Satish S (2007) Antimicrobial activity of some important medicinal plant against
plant and human pathogen. J Agri Sci 4
3. Srinivasan D, Nathan S, Suresh T (2007) Antimicrobial of certain Indian medicinal plants used
in folkloric medicine. J Ethnopharmacol 74:217–220
4. Raut S, Sen SK, Satpathy S, Pattnaik D (2012) An ethnobotanical survey of medicinal plants
inSemiliguda of Koraput District, Odisha, India. Bot Res Intern 5(4):97–107
5. Tan AC, Konczak I, Sze DM, Ramzan I (2010) Towards the discovery of novel phytochemicals
for disease prevention from native Australian plants: an ethnobotanical approach. Asian Pac J
Clin Nutr 19(3):330–334
6. Chakraborty RDB, Devanna N, Sen S (2012) North-East India an ethnic storehouse of unex-
plored medicinal plants. Scho Res Lib 2:143–152
7. Majumder J, Battacharjee PP, Datta BK, Agarwala BK (2014) Ethno-medicinal plants used by
Bengali communities in Tripura, Northeast India. J For Res 25:713–716
8. Semy K, Singh MR (2021) Quality assessment of Tsurang River water affected by coal mining
along the Tsurangkong Range, Nagaland, India. Appl Water Sci 11:115
Ethnobotanical Study of Medicinal Herbs Used by the Naga Tribes of Eastern Himalayas 23
9. Taid TC, Rajkhowa RC, Kalita JC (2014) A study on the medicinal plants used by the local
traditional healers of Dhemaji district, Assam, India for curing reproductive health related
disorders. Adv Appl Sci Res 5(1):296–301
10. Bharali P, Sharma CL, Singh B, Sharma M (2017) Ethnobotanical studies of spice and condi-
ment plants used by some communities of Assam. Int J of Adv in Sci Res 3(01):1–11
11. Sharma M, Das B (2018) Medicinal plants of north-east region of India: a small review. Int J
Curr Pharm Res 10(4):11–12
12. Chakre L, Narasimhan D (2013) Ethnobotany of Mao-Naga tribe of Manipur, India. Pleione
7(2):314–324
13. Usharani L, Singh WR, Surodhani S (2015) An ethnomedicinal plant-A less known Spices
used by Meitei Community of Manipur. Asian J Plant Sci Res 5(6):84–87
14. Khongsai M, Saikia SP, Kayang H (2011) Ethnomedicinal plants used by different tribes of
Arunachal Pradesh. Indian J Trad Know 10(3):541–546
15. Perme N, Choudhury SN, Choudhury R, Natung T, De B (2015) Medicinal plants in traditional
use at Arunachal Pradesh, India. Int J Phytopharm 5(5):86–98
16. Tripathi AK, Shankar R, Limasenla, Neyaz S (2016) Medicinal plants of Arunachal Pradesh
used in treatment of various diseases
17. Kashung S, Gajurel PR, Singh B (2020) Ethnobotanical uses and socio-economic importance
of climbing species in Arunachal Pradesh, India. Plant Sci Today 7(3):371–377
18. Deorani SC, Sharma GD (2007) Medicinal plants of Nagaland. In: Singh B., Singh M.P (eds),
Dehra Dun
19. Changkija S, Ajungla L, Rongsensashi, Mozhui R (2010) Medicinal and Aromatic Flora of
Nagaland. MPDA, Dept. of Forest, Ecology, Environment and Wildlife, Govt. of Nagaland
20. Lokho A (2012) The folk medicinal plants of the Mao Naga in Manipur, North East India. Int
J Sci Res Pub 2(6):1–8
21. Shankar R, Devalla RB (2012) Conservation of folk hailing practices and commercial medici-
nal plants with special reference to Nagaland. Int J Biodivers Conserv 4(3):155–163
22. Rongsensashi, Mozhui R, Changkija S (2013) Limasenla.: medicinal plants diversity of Fakim
wildlife sanctuary, Nagaland, India. Pleione 7(1):110–126
23. De LC (2016) Medicinal and aromatic plants of Northeast India. Int J Dev Res
06(11):10104–10114
24. Shimray RA, Lunleng A (2017) Ethnomedicinal knowledge of plants among the Tangkhul
Nagas of Manipur. Ind J Res Anthro 3(1):29–36
25. Jain SK, Rao RR (1977) A handbook of field and herbarium methods. Today and tomorrow
printers and publishers, New Delhi, p 157
26. Kanjilal UN, Kanjilal PC, Das A, Purkayastha C (1934) Flora of Assam, Ranunculaceae to
Elaeocarpaceae, vol 1. Government of Assam, Shillong, p 184
27. Bennet SSR (1987) Name changes in flowering plants of India and adjacent regions. Triseas
Publishers, Dehradun, p 772
28. Dey S (2018) Studies of the diversity of flowering plants of Tuensang district, Nagaland.
Ph.D. thesis, Nagaland University, Nagaland
Ethnomedicinal Use Reports of Seeds
as Tapped from Herbal Vendors in North
Maharashtra, India
1 Introduction
The word ‘Ethnobotany’ was almost unheard in India in the middle of last century.
‘Society of Ethnobotanists’ is a premier society in India established in 1980, which
filliped ethnobotanical studies particularly in India. This activity has well mush-
roomed and extended over nearly all Indian states and geographical regions inclu-
sive of majority of tribes and rural folks. It is needless to state that, throughout the
history of mankind, plants have been a major source of medicine. This central func-
tion still is continued worldwide particularly in the regions dominated by the tribal
communities and rural folks. The chief value of medicine in commerce in India was
stressed by Drury [1]. Prospects on ethnomedicinal studies in India are still bright
on account of remarkable phytodiversity and multiethnic culture inhabiting various
phytogeographical regions. Such investigations offer tremendous scope for bio-
prospecting in view of the sustainable development. Even today, medicinal sources
are sold in public places by herbal vendors in India. However, their traditional
knowledge is hardly documented yet. Probably, Sinha (1996) [2] is the first investi-
gator to tap their wisdom as an exclusive topic of research particularly in and around
Delhi. The present authors, therefore, extended inventory to document this much
neglected area of research in India, the results of which are being published [3–7].
This communication presents hitherto unprojected source of seeds sold by vendors
in northern part of Maharashtra (India).
Y. A. Ahirrao
S.S.V.P. Sanstha’s Arts, Commerce and Science College, Shindkheda,
Dhule, Maharashtra, India
M. V. Patil · D. A. Patil (*)
Department of Botany, S.S.V.P.Sanstha’s, L.K.Dr.P.R. Ghogrey Science College,
Dhule, Maharashtra, India
2 Methodology
The area under present study included five districts, namely, Nasik, Dhule,
Nandurbar, Jalgaon and Buldhana in northern part of Maharashtra state (India)
(Maps 1 and 2). The herbal vendors conduct their traditional family business of sell-
ing botanicals of medicinal importance. They carry on their business in public
places such as railway stations, bus stands, courts, Government offices, highways,
crossroads, chowks of cities, pilgrim centres, temples and places of weekly bazar
days. The vendors were interviewed and their medicinal wisdom like common plant
name, part or product used, diseases treated, doses, recipes and their administration,
age and sex of the patients to be treated, etc., were recorded. The samples were also
purchased and photographed. Botanical identities deciphered consulting various
floras like Hooker, 1872–1897; Cooke, 1958; Naik, 1998; Lakshminarsimhan and
Sharma, 1951; Patil, 2003; Kshirsagar and Patil, 2008; Diwakar and Sharma, 2002,
etc. [8–14] The information obtained is presented in Table 1 with necessary details.
3 Results
The present inventory on medicinal use reports of seeds from herbal vendors in
some districts of North Maharashtra. It revealed 101 species pertaining to 101 gen-
era and 48 angiospermic families. Majority of them are dicotyledons (94 species, 94
genera and 42 families), whereas monocotyledons contributed for a lesser share (07
species, 07 genera and 06 families). Out of total 101 taxa, 38 species are found to be
exclusively under cultivation, whereas 51 species are exclusively wild. Few others
(12 species) are either wild or cultivated as well. Of these, 27 species exhibited tree
habit, others being shrubs, herbs or climbers. Cultigens are also traditionally useful
Table 1 Seeds employed for treating human diseases
28
children
18. Butea monosperma (Lamk.) Palas Tree W D Powder Stomach worm One teaspoon Seven days
Taub. Paste Itching of skin twice a day Seven days
Fabaceae
(continued)
29
Table 1 (continued)
30
37. Drypetes roxburghii (Wall.) Jivan putra Tree C D Powder Sperm count One teaspoon Seven days
Huru. twice a day
Euphorbiaceae
(continued)
31
Table 1 (continued)
32
Euphorbiaceae powder
55. Lagenaria siceraria Dhudhi- Climber C D Paste Sleeplessness – Ten nights
(Molina) Standl. Bhopala Paste
Cucurbitaceae
(continued)
33
Table 1 (continued)
34
72. Ocimum tenuiflorum L. Krishna Tulas Shrub C D Seeds Malarial fever Twice a day Three to four
Lamiaceae days
(continued)
35
Table 1 (continued)
36
91. Terminalia chebula Retz. Hirada Tree C D Decoction Abdominal pain One cup twice Seven days
Combretaceae a day
(continued)
37
Table 1 (continued)
38
for various miscellaneous purposes, e.g. food, spice, ornamental, oil, shade tree,
beauty care, etc. in this region. The 101 species yielding seeds are employed for
various 74 human diseases (Table 1). They are administered in various forms of
medicinal recipes: (i) powder (72), (ii) paste (25), (iii) decoction (20), (iv) slurry
(03), (v) pellates (03), (vi) extract (03), (vii) ash (02), (viii) infusion (02), (ix) pulp
(01) and (x) seeds (01). Thus powder is the most common recipes advised by the
herbal vendors. Others are important in descending order presented here. The fig-
ures in parenthesis denote number of use-reports.
4 Discussion
Plants form the basis for most life on earth; however, the field of ‘Botany’ has
declined worldwide, while the field of ‘Ethnobotany’ keeps emerging. In this latter
missionary activity, India stands at the forefront of nations devoting sustained sci-
entific attention to this discipline of natural science. India has also recognized the
danger of the loss of precious traditional knowledge for the benefit of all humanity.
Some neglected areas of research in ethnobotany and ethnomedicine are being paid
serious attention [15]. One such study is tapping wisdom of herbal vendors who
conduct traditional business of selling botanicals of medicinal significance in public
places. The review of literature indicated that exclusive ethnomedicinal use reports
on specific plant parts are rare in India [7, 15–18]. Mahekar and Yadav [19–22]
informed about their ethnomedicinal utilities in Maharashtra as obtained from
herbal vendors. Recently, there are also few exclusive reports on ethnomedicine by
Patel and Parekh [23] and Kalim et al [24]. The present authors worked on this line
of research, the results of which are being published [3–7]. This communication is
an attempt to shed more light particularly on ethnomedicinal importance of seeds.
The Indian researchers do not want to ignore the contribution of native biore-
sources. The vast role played by seeds as cereals, millets, pseudo-cereals, oils,
spices and condiments is obvious for sustenance of human life since the appearance
of mankind on the Blue Planet. The panorama of ‘Ethnobotany’ has now been quite
widened in India [25]. However, its role in medicine is hardly emphasized [26–28].
Although the importance of newer subjects such as biotechnology and molecular
biology is being emphasized, there is equal necessity to nurture traditional subjects
like ‘Ethnobotany’ vis-à-vis ‘Ethnomedicine‘. The aforesaid new subjects help pro-
mote the traditional subjects for searching new lead molecules and propagation of
bioresources besides value addition and sustainable use of Indian phytodiversity. It
is equally important to tap traditional ethnomedicinal wisdom of Indian herbal ven-
dors for the welfare of mankind.
Acknowledgements The authors are thankful to the authorities of SSVP Sanstha for laboratory
and library facilities.
Ethnomedicinal Use Reports of Seeds as Tapped from Herbal Vendors in North… 41
References
1. Drury CH (1873) The useful plants of India with notices of their chief value in commerce,
medicine and the arts. William H. Allen & Co., London, UK
2. Sinha R (1996) Ethnobotany: the renaissance of traditional herbal medicine. INA Shree
Publishers, Jaipur
3. Ahirrao YA, Patil MV, Patil DA (2015) Traditional sources of antidotes from botanicals sold by
herbal vendors in North Maharashtra (India). Acad Res 10(1):156–160
4. Ahirrao YA, Patil MV, Patil DA (2015) Botanicals sold by herbal vendors employed for skin
diseases in North Maharashtra, India. Species 13(37):1–5
5. Ahirrao YA, Patil MV, Patil DA (2016) Ethnomedicinal investigation of herbal vendors in
North Maharashtra (India) combating jaundice disease. IRMJCR (Scholar World) Special
Issue VI(January, 2016):152–155
6. Ahirrao YA, Patil DA (2018) Ethnomedicinal investigation of some common botanicals sold
by vendors in North Maharashtra (India). Res World IX(Special Issue):175–180
7. Ahirrao YA, Patil MV, Patil DA (2021) Ethnomedicinal uses of flowers enquired from herbal
vendors in North Maharashtra (Maharashtra) India. PARIPEX-Indian J Res 10(11):1–4.
https://doi.org/10.36106/paripex
8. Hooker JD (1872–1897) The Flora of British India, vol. I–III. L.Reeve & Co., London, UK
9. Cooke T (1958) The Flora of The Presidency of Bombay, vol I–II. Bot.Surv.India, Calcutta
10. Naik VN (1998) Flora of Maharashtra, vol I-II. Amrut Prakashan, Aurangabad (M.S.)
11. Lakshminarsimhan P, Sharma BD (1991) Flora of Nashik District. B.S.I, Calcutta
12. Patil DA (2003) Flora of Dhule and Nandurbar District (Maharashtra). Bishen Singh Mahendra
Pal Singh, Dehradun
13. Kshirsagar SR, Patil DA (2008) Flora of Jalgaon District (Maharashtra). Bishen Singh
Mahendra Pal Singh, Dehradun
14. Diwakar PG, Sharma BD (2000) Flora of Buldhana District. Bot.Surv. India, Calcutta
15. Jain SK, Mugdal V, Banerjee DK, Guha A, Pal DC, Das D (1984) Bot. Surv. India, Howrah
16. Jain SK (1991) Dictionary of Indian Folk Medicine and Ethnobotany. Deep Publications,
New Delhi
17. Maheshwari JK (1996) Ethnobotany in South Asia. Scientific Publishers, Jodhpur
18. Goel AK, Tripathi S (2009) Ethnobotanical spectrum of Indian flora: An overview during the
past 20 years. J Ethnobot 21(1–2):131–153
19. Mahekar P, Yadav SR (2004) Botanical identity of some tubers and rhizomes from south west-
ern Maharashtra used in folk medicines. In: Janarthanam MK, Narsimhan D (eds) Plant diver-
sity, human welfare and conservation. Goa University Publication, Goa, pp 204–218
20. Mahekar P, Yadav SR (2004) Correct identity of some barks used in folk medicines. In: Ghate
et al (eds) Focus on sacred groves and ethnobotany. Prism Publications, Mumbai, pp 237–253
21. Mahekar P, Yadav SR (2004) Correct identity of some folk medicines of South Western
Maharashtra. Bull Bot Surv India 46/9(1–4):300–324
22. Mahekar P, Yadav SR (2008) Fruit and seed drugs used in folk medicines in South Western
Maharashtra. In: Chaturvedi A (ed) Ethnobotany & taxonomy of angiosperms. Rashtrasant
Tukadoji Maharaj Nagpur University, Nagpur (Maharashtra), pp 187–202
23. Patel PK, Parekh PP (2013) Therapeutic uses of some seeds among the tribals of Banaskantha
district, Gujarat, India. Rom J Biol Plant Biol 58(1):79–82
24. Kalim A, Zahreer M, Uddin M, Siddiqui A, Ahmed S, Hassan MM (2021) Nutritional
value, ethnomedicine, phytochemistry and pharmacology of Vigna radiat (L.) R.Wilczek. J
Pharmacogn Phytochem 10(2):54–58
25. Jain SK (2019) The widening panorama of ethnobotany in India. J Indian Bot Soc
98(3–4):98–102
26. Patil DA (2010) Medicinal Plants: History, Culture and Usage. Manglam Publications, Delhi
27. Patil DA (2019) Food Crops: Evolution, Diversity And Advances. Scientific Publishers, Jodhpur
28. Patil DA, Dhale DA (2013) Spices And Condiments: Origin, History And Applications. Daya
Publishing House (A Division of Astral International Pvt. Ltd.), New Delhi
An Ethnobotanical Study of Medicinal
Plants Used by Traditional Healers
in Grizzled Squirrel Wildlife Sanctuary
(GSWS) Tamil Nadu, India
Abbreviations
FC Frequent citation
Fic Informant consensus factor
FL Fidelity level
GSWS Grizzled squirrel wildlife sanctuary
RFC Relative frequency of citation
UV Use value
1 Introduction
Ethnobotany is the relationship and dealing between people and plants with respect
to their cultural values. Interactions and relationship between people and plants are
different from place to place because of their relative importance, uses and different
social, ethnic and cultural factors. Cultural values of plant exploration play a key
role in pharmaceutical and nutritional industrial sectors [1]. Ethnobotanists are
growingly focusing on the function of diverse quantitative and statistical techniques
to understand and gather knowledge on precious plants in induced communities [2].
Ethnobotany and ethnopharmacological knowledge is considered to be an integral
part of the knowledge required for drug development. Traditional medicine is to be
implicit as the sum total of the knowledge, talents and practices based on theories,
principles and understandings to different cultures that are employed to maintain
and improve health, as well as to prevent, diagnose and treat physical and mental
illnesses [3].
The World Health Organization (WHO) has a keen interest in documenting the
use of medicinal plants by native people from different parts of the world [4]
WHO further estimates that about 80% of the world’s population is dependent on
traditional medical practices for some aspect of primary health care [5]. According
to the FAO, in the last few decades, the number of known medicinal plants then
reaches up to 50,000 different species, which is 18.9% of the total world flora [6].
Still, valid scientific data on the practice of ethnomedicinal plants are moderately
incomprehensible. Recently, it is experimentally proved that plants offer immense
scope for researchers engaged in validation of traditional claims for the develop-
ment of novel drugs [7]. Since interest in traditional medicine has been increasing
world over, ethnobotanical studies have gained prominence to explore the tradi-
tional knowledge particularly in developing countries [8]. Therefore, collection of
ethnobotanical information and documentation of traditional knowledge have
gained prominence from the future prospective of drug development [9].
Conventional knowledge and applications have been marginalized due to political
and socio-economical grounds. Lately, interest in traditional medicine has been
initiated to explore the knowledge from various tribal groups across the country
[10–14]. Quite a few studies have related that tribal population in remote area, not
only rely on plant based resources for food, scavenge, medicines and fuel, but also
play a critical role in the administration of natural resources [8, 15–21]. A few
reports on ethnomedicinal uses of plants in the forests of Virudhunagar hills and
its adjoining areas were available [22–29], and most of the studies were conducted
qualitatively with a lacuna in data analysis. The present study was initiated with
an aim to identify knowledgeable resource persons among the Paliyar tribes in
Grizzled Squirrel Wildlife Sanctuary (GSWS), of Southern Western Ghats in
Tamil Nadu, India, and quantitatively analyze their indigenous ethnomedicinal
knowledge through various ethnobotanical tools on the utilization of commonly
used medicinal plants.
The Paliyars belong to the Southern tribal zone. The Southern tribes are historically
more ancient tribe. There are as many as 36 types of Scheduled Tribes in Tamil
Nadu. In the serialized list notified by the Government of Tamil Nadu, the Paliyars
are placed at the 32nd position. They live in the low-altitude regions of the Western
Ghats and live in large number in the study area (Virudhunagar district and parts of
Madurai district). Out of the total population of the scheduled tribes in Tamil Nadu
(7.21 lakh), the Paliyar tribe accounts for 2294. This is according to the census of
India in 2011 and Paliyar tribe ranks 20th among the tribal population. The Paliyars
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 45
can be grouped into three categories based on their life style, namely (i) nomadic
Paliyars, the hunter gatherers, who live in rock shelters wandering in tracts of the
forest in search of food and non-timber forests product; (ii) semi nomadic Paliyars,
also the hunter gatherers, who build hut and do not practice agriculture, and go out
to collect food and non-timber forest products and return to their dwelling and (iii)
settled Paliyars, who have land holdings, practicing agriculture and living mainly in
Kerala [30].
The Paliyars of the study area at present could be called semi nomadic type. Their
ancestors were nomadics. They live as individual families. Like other primitive
tribes, the Paliyars are short, dark complexioned, curly haired with thick protruding
lips and blunt nose with wide nostrils. Paliyars do not have any established mode of
dress (Fig. 1a,b). They are scantily dressed but freely wear whatever clothes are
available to them. They are non-vegetarians; however, they refrain from beef as
rigidly as the most orthodox Hindus [31]. Several species of Dioscorea provide the
basis of Paliyars staple food. Besides there are a wide variety of greens, stems,
tubers, unripe fruits and ripe fruits, which serve as alternative for food. Paliyars also
feed on wild animals and birds like rabbit, rat, deer, hen, etc.
The Paliyars live in small parties as isolated groups. Generally, a hamlet has
about 20 huts. Their small huts are exceptional with the walls prepared of mud or
with wiry intertwined stems of Lantana camara. Each hut is thatched with the
fronds of Cymbopogon citratus or Cymbopogon polyneuros or with the leaves of
Phoenix pusilla or Cocos nucifera (Fig. 1c). They sleep on mats, woven with the
leaves of the above said taxa. The Paliyars as a tribe do not possess much cohe-
siveness. Each settlement has its headman whose ability is never challenged, and
he is exclusively responsible for settling quarrels among the tribal people. They
are illiterate, but in recent years, they have started sending their children to nearby
schools.
The Paliyars are adroit in collecting honey. They collect honey from the branches
of towering tall trees and rock caves skilfully using special techniques. Paliyars
from four to seventy years old are agile on the trees. They also gather Kungillium
(resin) from the barks of Canarium strictum. They are also good hunters and they
trap deer, pig, boar, hare, wild fowl and flying squirrel. The meat obtained is divided
among the families within the settlement. They are good herbalists and often collect
medicinal plants from the forests. The knowledge about medicinal plant is rather
specialized and is limited to a few members in the community who are recognized
as “Vaidyars” or medicine men. They are normally respected the most and deemed
indispensable members inside the tribal society. Each medicine man cures all kinds
of illness, but some of them are concentrated in precise diseases. The remedies of
ordinary ailments such as cuts, fever, pain, headache and dysentery are recognized
by most members of the tribal society. There are several individuals in each locality,
46 P. S. Tresina et al.
Fig. 1 Paliyar tribes of area survey. (a) An old Paliyar tribal man of Athikoil sector of Grizzled
Squirrel Wildlife Sanctuary. (b) A tribal girl of Shenbagathoppu wearing a necklace made from the
seeds of Symplocoscochinchinensis. (c) A young tribal woman of Valliammal Nagar with her
children
who though not recognized as medicine men, possess such knowledge and act as
reliable informants. They maintain secrecy about the use of certain medicines
because they believe that the herbals will lose their healing power if too many peo-
ple know about them. Some practitioners have acceded to the knowledge of con-
vinced extraordinary remedies.
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 47
Paliyars have strong faith in religious conventions and practices. They habitually
practice Hindu religion. The deity of their worship is explicit to their region. The
Paliyars in the select study area worship Lord Siva and Ayyanar, a village god. They
also worship ‘Forest Goddess’ and Goddess ‘Poomadevi’ (Goddess Earth). On spe-
cial occasions, they stay in the temples in groups for 2 days and worship by offering
goats as sacrifice. On these days, a large quantity of Mullvallikizhangu (Dioscorea
pentaphylla var. pentaphylla) are collected and offered to the Goddess Earth
‘Poomadevi’.
Paliyars form a tribal group, and they are settled in the reserve forest area of the
Grizzled Squirrel Wildlife Sanctuary located in the South-Eastern slopes of Western
Ghats, Tamil Nadu, India. This wildlife sanctuary lies between 9°35′ and 9°8′ north
latitude and 77°4′ and 77°9′ east longitude (Fig. 2). It encompasses an area of 480
sq. km and is lying mostly in the district Virudhunagar and partly in Madurai. The
sanctuary was instituted in the year 1989. The attitude ranges from 100 to 2210 m
(MSL). It receives rainfall during the South-West as well as North-East monsoons.
The diverse climatic and topographic conditions exist in the sanctuary near a nota-
ble diversity of both the flora and fauna. The study area consists of tropical ever-
green forest, semi-evergreen forests, dry teak forests, southern mixed deciduous
forests and dry grasslands (Fig. 3). The study was conducted in 10 villages of GSWS
(Saduragiri, Thanniparai, Vandipannai, Nellikka Kottam, Valliammal Nagar,
Saragupannai, Petchikeni Kottam, Athikoil, Shenbagapthoppu and Ayyanarkoil),
which are inhabited by Paliyar tribes each consisting of 15–62 families distributed
in the deep forest areas.
48 P. S. Tresina et al.
Fig. 2 Location map of GSWS in Tamil Nadu, South-Eastern slopes of Western Ghats
The study area was investigated to get information from the tribal practitioners and
also to cross check the information provided by the other tribal practitioner during
the earlier visits. During each field trip, at least 10 days were spent with the local
people in their tribal hamlets. In order to document the utilization of medicinal
plants, field trips were made during 24 months period (July 2017 – June 2019)
ensuring that the dry and monsoon seasons were accommodated. A total of 12
resource persons or informants or traditional healers were identified to get the eth-
nomedicinal information through direct/interviews/oral conversations. They have
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 49
Fig. 3 Area surveyed. (a) The South Eastern slopes of Western Ghats, Ayyanarkoil. (b)
Shenbagathoppu Hills
50 P. S. Tresina et al.
Fig. 4 Format of field datasheet used to record the plant details with ethnomedicinal information
sound knowledge on the medicinal plants found in their surrounding areas and they
practice medicine within their families and neighbours. A field datasheet has been
prepared to record the plant details with ethnomedicinal information gathered from
the traditional healers (Fig. 4). Information on local name of plants, plant pieces
used for curing, technique of preparation, other plants/agents employed as ingredi-
ents, and modes of direction, etc., were recorded for each gathered ethnomedici-
nal plants.
The standard method of plant collection was followed, i.e. mounting, drying, prepa-
ration and conservation of plant specimens [32]. Coupon specimens of medicinal
plants in triplicate were gathered, organized and acknowledged. Plants with their
correct names were arranged alphabetically by family name, vernacular name, eth-
nomedicinal uses and other applications. The identification and nomenclature of the
assembled plants were supported by the Flora of Presidency of Madras [33] and the
Flora of Tamil Nadu Carnatic [34]. They were afterward confirmed at Botanical
Survey of India, Southern circle, Coimbatore, India. All the preserved herbarium
specimens were deposited at the herbarium of Ethnopharmacology Unit (EPH),
Research Department of Botany, V.O. Chidambaram College, Thoothukudi, Tamil
Nadu, India.
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 51
Based on the information obtained from the traditional healers in the study area, all
the reported ailments were categorized into 16 categories (Table 1), viz. gastro-
intestinal ailments (GIA), respiratory systems diseases (RSD), dermatological
infections/diseases (DID), genito-urinary ailments (GUA), skeleto-muscular system
disorders (SMSD), poisonous bites (PB), circulatory system/cardiovascular dis-
eases (CSCD), fever (Fvr), endocrinal disorders (ED), liver problems (LP), dental
care (DC), hair care (HC), ear, nose, throat problems (ENT), cooling agents (CA)
oncology (ONC) and general health (GH). Numerous diseases were placed in one
ailment group based on the body systems taken care of.
The informant consensus factor (Fic) was employed to see if there was concurrence
in the usage of plants in the ailment categories connecting the plant users in the
study area. The Fic was computed using the following formula [35]
N ur N t
Fic
N ur 1
where Nur refers to the number of use reports for a particular ailment category and
Nt refers to the number of taxa used for a particular ailment category by all infor-
mants. The product of this factor ranges from 0 to 1. A high rate (close to 1.0) des-
ignates that quite few taxa are used by a large percentage of the informants. A low
value designates that the informants disagree on the taxa to be employed in the
treatment inside a category of illness.
U
UV
n
where UV is the use value of a species, ‘U’ is the number of use reports cited by
each informant for a given plant species and ‘n’ is the total number of informants
interviewed for a given plant. UV is helpful in determining the plants with the
Table 1 Ailments grouped by different ailment categories
52
(continued)
53
Table 1 (continued)
54
highest use (most frequently indicated) in the treatment of an ailment. UVs are high
when there are many use reports for a plant and low when there are few reports
related to its use.
To determine the most frequently used plant species for treating a particular ailment
category by the informants of the study area, the fidelity level (FL) was calculated.
The FL was calculated using the following formula [37]
Np
FL % 100
N
where ‘Np’ is the number of use reports cited for a given species for a particular ail-
ment category and ‘N’ is the total number of use reports cited for any given species.
Generally, high FLs are obtained for plants for which almost all use reports refer to
the same way of using it, whereas low FLs are obtained for plants that are used for
many different purposes [38].
RFC FC / N 0 RFC 1
This index proves the local significance of each species, and it is specified by the
regularity of citation (FC, the number of informants mentioning the exercise of the
species) divided by the total number of informants participating in the survey (N),
without considering the used categories [39].
In the present study, 239 plants (224 plants collected and 15 plant parts/extracts
procured) belonging to 190 genera and 79 families were recorded, which were com-
monly used by most of the Paliyar traditional healers for the treatment of 122 types
of ailments. Of the recorded plants, 4 plants species belong to Pteridophytes and the
remaining 235 plants belong to the families of Angiosperms. Twelve informants
58 P. S. Tresina et al.
were selected from the study area based on their sound knowledge and long-term
experience of the use of medicinal plants. Information about the botanical name of
the plant, family, voucher specimen number, local (Tamil) name, life form, parts
used, mode of preparation, mode of administration, ailments treated, use reports,
use value and relative frequency citation are provided in Table 2. The prominent
family was Euphorbiaceae with 14 species, followed by Fabaceae, Apiaceae and
Lamiaceae with each 10 species, respectively (Fig. 5). Similarly, Yabesh et al. [18]
reported that the Euphorbiaceae family had the highest number of plant species
from a study carried out in Silent valley of Kerala.
The medicinal uses of plants gathered in the present study were compared with
the previously published information from other parts of India. It was found that
there were 44 claims from the plants such as Abrus precatorius, Acorus calamus,
Achyranthes aspera, Aloe vera, Allium sativum, Anisomeles malabarica, Asparagus
racemosus, Azadirachta indica, Acalypha fruticosa, Acalypha indica, Centella asi-
atica, Clitoria ternatea, Cardiospermum halicacabum, Capparis zeylanica,
Caralluma adscendens var. attenuata, Cocculus hirsutus, Bischofia javanica,
Evolvulus alsinoides, Euphorbia hirta, Gmelina asiatica, Ficus benghalensis,
Helicteres isora, Holopteles integrifolia, Leucus aspera, Lawsonia inermis, Lobelia
nicotianifolia var. nicotianifolia, Hybanthus enneaspermus, Mimosa pudica,
Morinda pubescens var. pubescens, Plumbago zeylanica, Plectranthus amboinicus,
Pergularia daemia, Pongamia pinnata, Phyllanthus emblica, Ocimum tenuiflorum,
Mimusops elengi, Mollugo pentaphylla, Rubia cordifolia, Sphaeranthus indicus,
Senna auriculata, Sida rhombifolia var. rhombifolia, Sterculia urens, Vitex negundo
and Ziziphus xylopyrus were reported for the first time from the study area. On the
other hand, no plants were reported as a fresh medicinal plant as all plants were
accounted with different uses.
The majority of the medicinal plants reported were herbs (47%) followed by trees
(23%), climbers (18%) and shrubs (12%) (Fig. 6). The high tradition of herbs in the
study area could be a signal of their abundance, and it might also be due to the real-
ity that they are effortlessly accessible and might have high effectiveness in the
treatment of ailments in comparison to other growth forms [40]. In addition to this,
herbs can be maneuvered with easiness in herbal grounding methods and drawing
out of bioactive compounds [41]. Concerning the use of the different plant parts, the
traditional healers commonly harvest the leaves (44%), whole plant, root, fruit
(10%), stem bark (9%), seed (8%), stem latex, tuber (5%), stem, unripe fruit, flower
(4%), aerial part and endosperm (3%) (Fig. 7). They were used for the preparation
of medicine solely or mixed with other plant parts. Many indigenous communities
elsewhere also utilized mostly leaves for the preparation of herbal medicines [5, 8,
Table 2 Synopsis of the commonly used medicinal plants by Paliyar tribes in GSWS
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
1 Abrus precatorius L. Kunnimuthu Cl Sd Paste Topical ENT 8 (Eye pain) 2.33 0.67
Fabaceae EPH 241 Rt Paste Oral SMSD 4 (Joint pain)
Lf Raw/paste Oral GUA2 (Improve sexual
chewed vigour)
Pb 6 (Poison bite)
DC 8 (Teeth sensitiveness)
2 Acacia columnaris Peiindu Cl Lf Paste Oral RSD 6 (Cold and cough) 0.50 0.33
Craib
Mimosaceae EPH
242
3 Acalypha fruticosa Sirusinni Sh Lf Paste/juice Topical/oral Pb 4 (Honey bee bite) 0.83 0.42
Forssk. SMSD 6 (Headache,
Euphorbiaceae EPH rickets)
243
4 Acalypha indica L. Kuppaimaeni He Lf Paste/juice Topical SMSD 4 (Headache) 1.00 0.50
Euphorbiaceae EPH DID 8 (Skin diseases)
258
5 Achyranthes aspera Nayurivi He Lf Paste/juice Oral Pb 4 (Dog bite) 1.50 0.75
L var. aspera DID 9 (Itches, skin
Amaranthacee EPH diseases)
261 GIA 5 (Dysentery)
6 Acorus calamus L. Vasambu He Rh Raw/juice Topical/oral SMSD12 (Sedative, 1.00 0.83
Araceae EPH 84 giddiness)
7 Actiniopteris radiata Mayilkalpul He Lf Paste Topical GIA 6 (Colic pain) 0.50 0.17
(Sw.) Link
Actinopteridacae
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
EPH 56
(continued)
59
Table 2 (continued)
60
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
8 Aegle marmelos (L.) Vilvam Tr Lf Paste Topical DID 4 (Itches) 1.92 0.75
Correa UFt Paste Oral GIA 10 (Diarrhoea)
Rutaceae EPH 138 Ft Juice Oral GIA 9 (Ulcer)
9 Albizia amara Usilai Tr Sb Paste Oral GIA 6 (Stomachache) 0.50 0.58
(Roxb.) Boivin.
Mimosaceae EPH
244
10 Allium cepa L. Vengayam He Bb Paste Topical SMSD11 (Rheumatism, 1.75 0.91
Alliaceae EPH 245 headache)
DID10 (Skin diseases)
11 Aloe vera (L.) Chotthukathalai He Lf Raw (gel) Oral/topical CA12 (Body cooling) 1.75 0.91
Burm. f. DID 9 (Eczema, wound
Liliaceae EPH 246 burn)
12 Alstonia scholaris Ezhilaipalai Tr Sb Paste Oral GUA 8 (Lactation) 1.42 0.42
(L.) R. Br RSD 9 (Asthma)
Apocynaceae EPH 259
EPH 247
(continued)
61
Table 2 (continued)
62
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
23 Aristolochia indica Thalaisurulivaer Cl Rt Paste/powder/ Topical/oral Pb 9 (Snake bite, scorpion 1.50 0.50
L. juice sting)
Aristolochiaceae SMSD 9 (Headache)
EPH 248 GIA 2 (Stomachache)
24 Aristolochia tagala Karudakkodi Cl Rt Paste/powder Topical/oral Pb 9 (Snake bite, scorpion 1.67 0.33
Cham. sting)
Aristolochiaceae SMSD 8 (Headache)
EPH 249 GIA 3 (Stomachache)
25 Asclepias Kuruthipoo He Lf Powder Oral DID 4 (Skin diseases) 1.08 0.42
curassavica L. Rt Decoction/paste Oral GIA 9 (Constipation, piles)
Asclepiadaceae
EPH 250
26 Asparagus Neervalli Cl Tu Boiled Oral GIA 4 (Bed wetting) 0.83 0.50
racemosus Willd. Lf Paste Topical Pb 6 (Honeybee sting)
Liliaceae EPH 251
27 Atalantia racemosa Kattuelumichai Tr Lf Decoction Topical DID 7 (Itches, skin 1.08 0.17
Wight & Arn. Ft Juice Oral diseases)
Rutaceae EPH 139 GIA 6 (Stomach disorder)
28 Azadirachta indica Vembu Tr Lf Boiled (vapour) Topical RSD 8 (Cold and cough) 2.50 0.83
A. Juss. Sb Decoction (inhalation) Fvr 10 (Fever)
Meliaceae EPH 140 Sd Juice Oral SMSD 12 (Rheumatism)
Tropical
29 Bambusa Mungil Tr Lf Juice/paste Oral/topical SMSD 4 (Sprain) 0.58 0.17
arundinacea (Retz.) RSD 3 (Chest pain)
Roxb.
Poaceae EPH 141
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
30 Basella alba L. var. Kattupasali Cl TU Paste Topical GIA 9 (Colic pain) 2.25 0.25
alba Lf Juice Oral CA 10 (Body cooling)
Chenopodiaceae St Infusion Oral GIA 8 (Laxative)
EPH 142
31 Bauhinia racemosa Aathi Tr Lf Juice Topical ENT 4 (Removal of dust 0.33 0.17
Lam. from eye)
Caesalpiniaceae
EPH 112
32 Bauhinia tomentosa Kanchini Sh Lf Infusion Oral CA 4 (Body cooling) 0.33 0.17
L.
Caesalpiniaceae
EPH 113
33 Begonia malabarica Narayana He Ap Paste Topical SMSD 11 (Rheumatism) 1.50 0.33
Lam. sanjivi Lf Raw/powder Oral RSD 7 (Astringent)
Begoniaceae EPH
114
34 Begonia picta Sm. Kalthamarai He Lf Raw Oral GH 7 (Refrigerant) 0.58 0.17
Begoniaceae
EPH 89
35 Benkara malabarica Kuthupidari Sh Sb Paste Topical Pb 6 (Scorpion sting) 0.50 0.17
(Lam.) Tirvengadam
Rubiaceae EPH 130
36 Bidens pilosa L. var. Kattu He Lf Juice Oral LP 6 (Jaundice) 0.50 0.25
minor (Blume) karisalankanni
Sherff
Asteraceae EPH 321
37 Bischofia javanica Omaviruchu Tr Sb Boiled Topical SMSD 8 (Body pain) 0.67 0.25
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
Blume (Filtrate)
Bischofiaceae EPH
63
322
(continued)
Table 2 (continued)
64
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
38 Blepharis Sadhaiotti He Lf Paste Topical DID 5 (Weepon. injuries, 1.83 0.50
maderaspatensis Sd Infusion Oral wound)
(L.) Heyne ex Roth GIA 11 (Ulcer)
Acanthaceae EPH GUA 6 (Urinary infection)
301
39 Blumea barbata Mayakkam He Lf Raw (aroma) Topical SMSD 6 (Giddiness) 0.50 0.17
DC. neeki patchilai (inhalation)
Asteraceae EPH 93
40 Bupleurum wightii Kattuseeragam He Ft Infusion Oral CA 7 (Body cooling) 0.58 0.17
Mukh var.
ramosissimum
(Wight & Arn)
Chandrabose comb.
nov.
Umbelliferae (nom.
alter. Apiaceae)
EPH 94
41 Caesalpinia crista L Kalachichedi Sh Sd Paste Topical SMSD10 (Hydrocele) 0.83 0.17
Caesalpiniacee
EPH 95
42 Calotropis gigantea Erukku Sh Sl Paste Topical RSD 9 (Cold and Cough) 1.92 0.83
(L.) R. Br. Rt Juice Topical SMSD 6 (Headache, body
Asclepiadaceae Lf Warmed Topical pain)
DPH 303 (poultice) SMSD 8 (Rheumatism)
43 Canarium strictum Kungiliam Tr Re Powder Topical SMSD 6 (Headache) 1.17 0.67
Roxb. (burned) (inhalation) RSD 8 (Cold)
Burseraceae EPH
305
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
44 Canscora perfoliata Narayana He Wp Juice Oral Pb 7 (Poison bite) 0.58 0.50
Lam. vembu
Gentianaceae EPH
308
45 Capparis sepiaria Muruvilikodi Sh Sb Paste Topical DID 7 (Eczema, dandraff) 1.08 0.42
L. Lf Paste Oral CA 6 (Body cooling)
Capparaceae
EPH 57
46 Capparis zeylanica Kaathatikaai Cl Lf Paste Topical SMSD 11 (Arthritis) 0.92 0.33
L.
Capparaceae
EPH 60
47 Capsicum annum L. Periya He Ft Paste Topical DC 4 (Toothache) 0.58 0.67
Solanaceae EPH 61 usimilagai ENT 3 (Sore throat)
48 Caralluma Periya He AP Raw Oral GH10 (Refrigerant, reduce 0.83 0.83
adscendens (Roxb.) sirumankeerai body weight)
Haw. var. attenauta
(Wight) Grav &
Mayuranathan
Asclepiadaceae
EPH 401
49 Caralluma lasiantha Sirumankeerai He AP Raw Oral GH10 (Refrigerant, reduce 0.83 0.75
(Wight) N.E.Br body weight)
Asclepiadaceae
EPH 402
50 Cardiospermum Mudakkathan Cl Lf Paste/juice Topical/oral SMSD12 (Rheumatism, 1.67 0.50
halicacabum L. Decoction Oral Giddiness)
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
Table 2 (continued)
66
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
51 Carmona retusa Siruthani Sh Wp Raw Topical (touch) GH 4 (Puberty) 0.33 0.17
(Vahl) Masamune
Boraginaceae EPH
100
52 Centella asiatica Vallarai He Lf Raw/juice Oral RSD 6 (Cold and cough, 0.50 0.67
(L.) Urban chest pain)
Umbelliferae (nom.
alter. Apiaceae)
EPH 412
53 Ceropegia juncea Thiralankodi Cl Sl Raw Topical Pb 3 (Insect bite) 0.58 0.17
Roxb. St Juice Oral GIA 4 (Anthelmintic)
Asclepiadaceae
EPH 413
54 Cheilanthes Poorankadi- He Rh & Rc Paste Topical Pb 4 (Scorpion sting) 0.33 0.17
opposita Kaulf chedi
Sinopteridaceae
EPH 415
55 Chionanthus Vandukadipattai Tr Sb Paste Topical DID 4 (Itches) 0.33 0.17
mala-elengi
(Dennst.) P.S.Green
Oleaceae EPH 27
56 Cissampelos pareira Malaithangivaer Cl Rt Powder Oral Pb 9 (Poison bite) 1.33 0.33
L. var. hirsuta GIA7 (Stomachache)
(Buch-Ham. DC)
Forman
Menispermaceae
EPH 78
57 Cissus Perandai Cl Lf Juice/paste Oral ONC 4 (Tumor Tumor-like 1.33 0.50
quadrangularis L. swelling)
P. S. Tresina et al.
(continued)
67
Table 2 (continued)
68
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
65 Commelina Amala He Ss Raw Topical ENT 10 (Removal of dust 0.83 0.83
benghalensis L. from the eyes)
Commelinaceae
EPH 368
66 Commelina ensifolia Amala He Ss Raw Topical ENT 10 (Removal of dust 0.83 0.83
R.Br. from the eyes)
Commelinaceae
67 Commiphora Mangkiluvai Tr Es Raw Oral GIA 7 (Stomachache) 0.58 0.17
caudata (Wight &
Arn.) Engler
Burseraceae EPH
371
68 Commiphoran Kodikiluvai Tr Es Raw Oral GIA 6 (Stomachache) 0.50 0.17
pubescens (Wight &
Am.) Engler
Burseraceae EPH
372
69 Coriandrum sativum Kothamalli He Lf Raw Oral GIA10 (Anthelmintic, 2.50 0.83
L. Ft Boiled Oral digestive stimulant, gastric
Umbelliferae (nom. complaints
alter. Apiaceae) GIA 12 (Indigestion)
EPH 341
70 Crataeva adansonii Mavulingam Tr Lf Boiled (Coconut Topical SMSD 8 Joint pain 1.00 0.25
DC. subsp. odora Sb oil) Oral SMSD 4 (Headache)
(Buch-Ham) Jacobs Decoction RSD 8 (Asthma, cough,
Capparaceae EPH bronchitis, skin diseases)
342
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
71 Croton Reilpoondu Ae Sl Raw Topical DID 4 (Thorn injury) 0.33 0.25
bonplandianum
Baill.
Euphorbiaceae EPH
343
72 Curcuma longa L. Manjal He Rh Roasted Oral GIA 8 (Bowel disorders) 1.00 0.67
Zingiberaceae EPH Powder Topical Pb 4 (Insect bite)
199 DID 10 (Antiseptic, skin
diseases)
73 Curculigo Kuluthupokie/ He Tu Powder Oral GUA 12 (Improve sexual 1.83 0.83
orchioides Gaertn. Nilapanai vigour)
Hypoxidaceae EPH
198
74 Cyanotis tuberosa Palvalikizhangu He Tu Roasted Topical DC 10 (Toothache) 0.83 0.25
(Roxb.) Schultes &
Schultes f.
Commelinaceae
EPH 200
75 Cymbopogon Taragupullu He Tu Roasted Topical SMSD 4 (Giddiness) 2.50 0.83
citratus (DC.) Stapf. Lf Juice (Inhalation) GIA10 (Stomachache,
Poaceae EPH 201 Oral dysentery, bowel
complaints, vomiting)
Fvr 8 (Fever)
SMSD 8 (Headache)
76 Cynodon dactylon Arugampullu He Lf Juice Oral CSCD 8 (Heart tonic) 3.08 0.91
(L.) Pers SMSD10 (Rheumatism)
Poaceae EPH 202 GIA 10 (Gastric
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
complaints, indigestion)
CA 9 (Body cooling)
69
(continued)
Table 2 (continued)
70
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
77 Dichrostachys Vidathalai Tr Lf Paste Oral GIA 5 (Dysentery) 0.42 0.25
cinerea (L.) Wight
& Arn
Mimosaceae EPH
214
78 Dioscorea bulbifera Vethalaivalli/ Cl Tu Raw/cooked Oral GIA 9 (Dysentery) 0.75 0.17
L. var. vera Prain & Karuvalli
Burkill
Dioscoreaceae EPH
215
79 Dioscorea Vethalaivalli Cl Tu Raw/cooked Oral GIA 9 (Dysentery) 0.75 0.17
oppositifolia L. var.
dukhumensis Prain
& Burkill
Dioscoreaceae EPH
218
80 Dioscorea Mullvalli Cl Tu Cooked/roasted Oral GIA 11 (Piles) 0.92 0.25
pentaphylla L. var.
pentaphylla
Dioscoreaceae EPH
220
81 Dioscorea Noolvalli Cl Tu Cooked Oral GIA 10 (Bowel disorders) 0.83 0.50
tomentosa Koen. ex
Spreng.
Dioscoreaceae EPH
221
82 Diplocyclos Malaipusanni Cl Tu Roasted Oral GUA 8 (Abortion) 0.67 0.67
palmatus (L.)
Jeffrey
P. S. Tresina et al.
Cucurbitaceae EPH
101
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
83 Dodonaea viscosa Virali Sh Lf Boiled Topical SMSD 9 (Body pain) 0.75 0.50
(L.) Jacq (oil) (massages)
Sapindaceae EPH
102
84 Drynaria quercifolia Mudavathukaal He Rh Paste Topical SMSD12 (Rheumatic pain, 1.00 0.25
(L.) J. Sm. bone fracture)
Drynariaceae EPH
103
85 Drypetes sepiaria Kalvirai Tr Sb Paste Topical DID 4 (Katti) 0.33 0.17
(Wight & Arn.) Pax
& Hoffm.
Euphorbiaceae EPH
104
86 Eclipta prostrata Karisalankanni He Lf/Wp Juice Topical (bath) HC 12 (Graying Greying 1.50 0.83
(L.) L. of hair, hair loss)
Asteraceae EPH 177 CA 6 (Body cooling)
87 Eleusine coracana Kattukepai He Gr Cooked Oral ED 7 (Diabetes) 0.58 0.83
(L.) Gaertn.
Poaceae EPH 178
88 Ensete superbum Malaivazhai Tr Pd Juice Oral GUA 11 (Kidney stone) 0.92 0.75
(Roxb.) Cheesman
Musaceae EPH 179
89 Eugenia discifera Vellainaaval Tr Sb Powder Oral GIA 5 (Stomachache) 0.42 0.17
Gamble
Myrtaceae EPH 456
90 Euphorbia hirta L Amanpatcharisi He Sl Raw Topical GUA 4 (Lactation) 1.33 0.33
Euphorbiaceae EPH Lf Warmed Topical SMSD 6 (Swelling)
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
Table 2 (continued)
72
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
91 Euphorbia rosea Amanpatcharisi He Sl Raw Topical GUA 6 (Lactation) 0.50 0.17
Retz.
Euphorbiaceae EPH
462
92 Evolvulus alsinoides Vishnukarandi He Lf Decoction Oral Fvr 12 (Fever) 2.25 0.67
(L.) L. Rt RSD 10 (Cold and cough)
Convolvulaceae Wp GUA 5 (Venereal diseases)
EPH 463
93 Ficus benghalensis Aal Tr Sl Raw Topical DID10 (Heal cracker) 1.83 0.50
L. var. benghalensis Sb Powder Topical (tooth DC 12 (Tooth strength)
Moraceae EPH 464 powder)
94 Ficus microcarpa L. Punniyavirusu Tr Sl Raw Topical DID 12 (Heal cracker) 1.00 0.50
f.
Moraceae EPH 465
95 Ficus racemosa L. Atthi Tr Sl/Lx Raw Topical DC 12 (Heal cracker boils, 2.33 0.50
Moraceae EPH 466 Sb Infusion Topical (mouth Blisters
Paste wash) DC 10 (Mouth wash)
Topical DID 6 (Burns)
96 Ficus religiosa L. Arasu Tr Sl Raw Topical DID 9 (Fissures in foot) 0.75 0.42
Moraceae EPH 467
97 Flacourtia indica Mullumayilai Sh Ft Raw/powder Oral RSD 5 (Asthma) 0.42 0.17
(Burm. f.) Merr. Uft
Flacourtiaceae EPH
468
98 Foeniculum vulgare Sombu He Ft Raw/powder Oral GIA 9 (Improve digestion) 0.75 0.42
Mill
Umbelliferae (nom.
Alter Apiaceae)
P. S. Tresina et al.
EPH 32
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
99 Gardenia resinifera Kattu koiya/ Tr Re Raw Oral RSD 6 (Cough) 0.50 0.17
Roth Vetchi Pc Powder
Rubiaceae EPH 457 Isd
100 Givotia Vandalai Tr Es Powder Oral GID 8 (Improve digestion) 0.67 0.17
rottleriformis Griff.
Euphorbiaceae EPH
458
101 Globba marantina Kattumanjal He Rh Paste Topical DID 3 (Katti) 0.25 0.17
L.
Zingiberaceae EPH
459
102 Gloriosa superba L. Karthika Cl Uft Boiled (coconut Topical ENT 12 (Ophthalmic 1.00 0.67
Liliaceae EPH 482 kilangu oil) problem)
103 Gmelina asiatica L Kumulu Sh Lf Infusion Oral CA 6 (Body cooling) 0.92 0.42
Verbenaceae EPH Ft Juice Topical ENT 5 (Sore throat)
483
104 Gymnema sylvestre Shirukurinjan Cl Lf Powder Oral ED 12 (Diabetes) 1.75 0.91
(Retz.) R. Br. Ex Wp Decoction Oral CSCD 9 (Cardiovascular
Schultes disorders, obesity, lowering
Asclepiadaceae cholesterol)
EPH 484
105 Gyrocarpus Tanakku Tr Es Raw Oral GIA 6 (Stomachache) 0.50 0.17
asiaticus Willd.
Hemandiaceae EPH
485
106 Hedyotis brachiata Inbural He Lf Juice Oral GIA 6 (Vomiting) 0.50 0.17
Wight
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
107 Hedyotis corymbosa Inbural He Lf Juice Oral GIA 7 (Vomiting) 0.58 0.33
(L.) Lam
Rubiaceae EPH 490
108 Hedyotis puberula Emburel He Lf Powder Oral ED 3 (Diabetes) 0.25 0.17
(G. Don) Arn.
Rubiaceae EPH 491
109 Helicteres isora L. Valamburi Sh Ft Ash Topical ENT 6 (Sore in ear) 1.50 0.50
Malvaceae EPH 492 Lf Paste Topical DID 5 (Skin diseases,
Rt Decoction Oral eczema, scabies)
RSD 7 (Asthma)
110 Hemidesmus indicus Nannari Cl Rt Powder Oral CA 12 (Body cooling) 2.83 0.83
(L.) R. Br. var. GIA 10 (Improve
indicus digestion)
Periplocaceae GH 12 (Thirstness)
EPH 62
111 Hemidesmus indicus Nannari Cl Rt Powder Oral CA 12 (Body cooling) 2.92 0.83
(L.) R. Br. var. GIA 11 (Improve
pubescens (Wight & digestion)
Arn) Hook.f GH 12 (Thirstness)
Periplocaceae
EPH 63
112 Heracleum rigens Kattu He Yp Subgy Oral GIA 3 (Bowel complaints) 0.25 0.17
Wall. ex DC. var. kothamalli
rigens
Umbelliferae (nom.
alter. Apiaceae)
EPH 45
113 Hibiscus vitifolius Periyathutthi He Fl Paste Topical DID 3 (Boils) 0.25 0.25
L.
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
120 Ipomoea Thoolikkodi Cl Lf Paste Topical DID 3 (Wounds) 0.25 0.17
barlerioides Benth.
ex Clarke
Convolvulaceae
EPH 521
121 Ipomoea hederifolia Neelikodi Cl Lf Paste Topical DID 5 (Wounds) 0.42 0.25
L.
Convolvulaceae
EPH 523
122 Ipomoea obscura Thazhiilai Cl Lf Paste Topical DID 6 (Boils) 0.50 0.25
(L.) Ker- Gawl.
Convolvulaceae
EPH 524
123 Ipomoea staphylina Onaan kodi Cl Sl Raw Topical DID 7 (Fissures in foot) 0.58 0.17
Roem. & Schultes
Convolvulaceae
EPH 525
124 Jasminum Kattumalli Cl Fl Boiled Topical ENT 6 (Oozing pus in ear) 0.92 0.17
roxburghianum Rt Paste Oral GIA 5 (Colic pain)
Wall.
Oleaceae EPH 555
125 Jatropha curcas L. Kattamanaku Tr Lf Juice Topical SMSD 6 (Rheumatism) 1.08 0.58
Euphorbiaceae EPH Rt Raw Topical GUA 7 (Abortion)
556
126 Jatropha Vellaiadalai Sh Wp Juice Topical SMSD 10 (Rheumatism) 0.83 0.50
glandulifera Roxb
Euphorbiaceae
EPH557
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
127 Jatropha Sivappuatalai Sh Wp Juice Topical SMSD 9 (Rheumatism) 0.75 0.50
gossypifolia L.
Euphorbiaceae EPH
558
128 Justicia adhatoda L. Adatoda Sh Lf Decoction Oral RSD12 (Asthma, cold and 1.00 0.75
Acanthaceae EPH cough, respiratory
151 problems)
129 Justicia glauca Rottl Neemotchi/ He Lf Juice Oral Pb 6 (Poison bite) 0.50 0.25
Acanthaceae EPH Kattukurinchi
152
130 Kalanchoe olivacea Kulirthamarai He Lf Raw Oral CA 10 (Body cooling) 0.83 0.17
Dalz. & Gibs.
Crassulaceae EPH
154
131 Kalanchoe pinnata Megasanjeevi He Lf Raw Oral CA 10 (Body cooling) 0.83 0.17
(Lam.) Pers.
Crassulaceae EPH
158
132 Kleinia grandiflora Musakkalli Sh Rt/Lf Boiled (coconut Topical SMSD 8 (Joint pain) 0.67 0.25
(Wall. ex DC) oil)/Paste (massage)
N. Rani. Topical
Asteraceae EPH 156
133 Knoxia sumatrensis Vazhukkanangai He Lf Powder Oral Pb 3 (Poison bite) 0.25 0.17
(Retz.) DC. var.
linearis
Rubiaceae EPH 157
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
77
Table 2 (continued)
78
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
134 Lantana camara L. Unnipoo Shrub Sh Fl Paste Topical Pb 4 (Insect bite) 0.33 0.25
var. aculeata (L.)
Mold
Verbenaceae EPH
146
135 Lantana wightiana Muraikaichal Sh Lf Paste Topical Fvr 5 (Fever) 0.42 0.25
Wall. ex Gamble chedi
Verbenaceae EPH
290
136 Launaea sarmentosa Sulukkupatchi- He Lf Paste Topical SMSD 6 (Sprain) 0.50 0.17
(Willd.) Sch-Bip.ex lai
Kuntze
Asteraceae EPH 641
137 Lawsonia inermis L. Marudondi Sb Lf Boiled (coconut Topical (head) HC12 (Hair growth, hair 1.83 0.67
Lythraceae EPH 44 oil) loss)
CA10 (Body cooling)
138 Lepisanthes Vaivumaram Tr Sb Paste Oral GIA 8 (Gastric complaints) 0.67 0.17
senegalensis (Juss.
ex Poir.) Leenh
Sapindaceae EPH
291
139 Leucas aspera Tumbai He Lf Juice Topical SMSD 10 (Migraine) 0.83 0.42
(Willd.) Link
Lamiaceae EPH 292
140 Leucas biflora Siru tumbai He Ap Boiled Topical SMSD 10 (Headache) 0.83 0.42
(Vahl) R. Br var. (inhalation)
biflora
Lamiaceae EPH 293
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
141 Limonia acidissima Vila Tr Lf Infusion Oral Fvr 6 (Fever) 0.50 0.17
L.
Rutaceae EPH 295
142 Lobelia Kattupugaiyilai He Lf Chawed Oral GH 3 (Fatigue) 0.25 0.17
nicotianifolia Roth
ex Schultes var.
nicotianifolia
Lobeliaceae EPH
298
143 Mangifera indica L. Maa Tr Es Powder Oral GIA 8 (Stomachache) 0.67 0.50
Anacardiaceae EPH
299
144 Memecylon edule Sarkarai vembu Tr Lf Powder Oral ED 8 (Diabetes) 0.67 0.83
Roxb.
Melastomataceae
EPH 216
145 Micromeria biflora Elumichai He Wp Infusion Topical HC 10 (Hair tonic, hair 0.83 0.58
(Buch.- Ham. ex vallarai growth, hair loss)
D. Don.) Benth.
Lamiaceae EPH 223
146 Mimosa pudica L. Thotalsinungi He LR/Wp Paste Topical DID 3 (Insect bite) 1.67 0.42
Mimosaceae EPH SMSD11 (Inflamed joints)
224
147 Mimusops elengi L. Mahilampoo Tr Sb Paste/juice Oral GUA 9 (Improve fertility 0.75 0.50
Sapotaceae EPH in women)
225
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
79
Table 2 (continued)
80
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
148 Mollugo cerviana Pappadai He Wp Decoction Oral RSD 9 (Cold and cough) 1.42 0.33
(L.) Ser. Paste Topical DID 8 (Eczema, bad body
Molluginaceae odour)
EPH 58
149 Mollugo pentaphylla Pappadai He Wp Infusion Oral GUA 7 (Menstrual 1.50 0.58
L. Roasted Topical discharge)
Molluginaceae Decoction Poultice DID 6 (Sore legs)
EPH 59 Oral ENT 5 (Eye diseases)
150 Momordica Kuruvithalai He Uft Cooked Oral GIA 10 (Anthelmintic) 1.83 0.83
charantia L. var. ED 12 ((Diabetes)
charantia
Cucurbitaceae
EPH 16
151 Morinda pubescens Manjanatti Tr Lf Infusion Oral GIA 6 (Digestion 1.83 0.83
J.E. Smith var. Sb Paste Topical disorders, piles)
pubescens Decoction Oral DID 4 (Weapon injury,
Rubiaceae DPH 560 leucoderma)
SMSD 12 (Rheumatism)
152 Moringa Kattumurungai Tr Lf Paste/juice Oral LP 6 (Jaundice) 1.67 0.58
concanensis Nimmo GIA 8 (Bowel disorder)
ex Gibs.
Moringaceae EPH
564
153 Mucuna Thellukkai Cl Sd Paste Topical SMSD 6 (Bone fracture) 0.50 0.58
atropurpurea DC.
Fabaceae EPH 570
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
154 Mukia Musumusukki Cl Lf Juice Oral RSD 10 (Asthma, cold and 2.33 0.67
maderaspatana (L.) Decoction Oral cough)
M. Roem. SMSD 12 (Rheumatism)
Cucurbitaceae EPH GUA 6 (Diuretic)
573
155 Mundulea sericea Vellaipurasu Tr Lf Paste Topical DID 6 (Leucoderma) 0.50 0.17
(Willd.) A. Cheval.
Fabaceae EPH 576
156 Murraya koenigii Kariveppilai Tr Lf Paste Topical HC 10 (Hair growth, hair 1.75 0.58
(L.) Spreng. Juice Oral loss, hair tonic)
Rutaceae EPH 66 ED 11 (Diabetes)
157 Nothopegia Karunthillai Tr Sd/Lf Powder Oral Pb 3 (Snake bite) 0.25 0.17
colebrookeana
(Wight) Blume
Anacardiaceae EPH
440
158 Ochna lanceolata Mayilai Tr Sb Powder/Juice Oral GIA 7 (Stomachache) 1.42 0.50
Spreng. GUA 10 (Abortion,
Ochnaceae EPH 443 menstrual disorder)
159 Ocimum tenuiflorum Tulsi He Lf Boiled (vapour) Topical SMSD 4 (Headache) 1.83 0.67
L. Paste (inhalation) DID 8 (Wounds)
Lamiaceae EPH 444 Raw Topical RSD 10 (Cold and cough)
Oral
160 Orthosiphon Kattutulsi He Lf Paste Topical DID 3 (Wounds) 0.25 0.17
thymiflorus (Roth)
Sleensen
Lamiaceae EPH 600
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
81
Table 2 (continued)
82
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
161 Parahemionitis Mayilkal He Lf Paste Topical GIA 8 (Colic pain) 0.67 0.17
cordata (Roxb. ex patchilai
Hook. & Grev.)
Fras. Jenk.
Hemionitidaceae
EPH 601
162 Pavonia odorata Thuthi He Lf Paste Topical DID 8 (Wounds) 0.67 0.17
Willd.
Malvaceae EPH 611
163 Pergularia daemia Velipparuthi Cl Lf Paste Topical GIA 10 (Gastric 1.83 0.75
(Forssk.) Chiov. Rt/Lf Boiled Topical complaints)
Asclepiadaceae (massage) SMSD 12 (Joint pain)
EPH 612
164 Phyllanthus amarus Kilanelli He Ap Juice Oral LP 12 (Jaundice) 1.00 0.91
Schum. & Thonn.
Euphorbiaceae
EPH 83
165 Phyllanthus emblica Nelli Tr Ft Juice Oral GUA 6 (Diuretic) 1.17 0.83
L. GIA 8 (Digestive disorder)
Euphorbiaceae
EPH 82
166 Phyllanthus Kilaneli He Ap Juice Oral LP 12 (Jaundice) 1.00 0.83
maderaspatensis L.
Euphorbiaceae
EPH 81
167 Physalis minima L. Toppipalam He Lf Cooked Oral GIA 10 (Ulcer) 0.83 0.58
Solanaceae EPH 71
P. S. Tresina et al.
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
168 Pimpinella Kothuvallarai He Wp Powder Oral GH 3 (Refrigerant) 0.25 0.42
candolleana Wight
& Arn.
Umbelliferae (nom.
alter. Apiaceae)
EPH 72
169 Piper argyrophyllum Kattumilagu Cl Lf/Ft Raw Oral RSD 10 (Cold and cough) 1.75 0.67
Miq. Decoction RSD 11 (Cold and cough)
Piperaceae EPH 77
170 Piper trioicum Roxb Vethalaikodi Cl Lf/St Boiled Topical (bath) ENT 7 (Buring sensation 0.58 0.50
Piperaceae EPH 18 in the eye)
171 Plectranthus Omavalli He Lf Juice Oral RSD 12 (Asthma, cold and 1.58 0.67
amboinicus (Lour.) cough, bronchitis)
Spreng. GIA 7 (Anthelmintic)
Lamiaceae EPH 445
172 Plectranthus Poolankilangu He Rh Paste Topical DID 4 (Katti) 0.33 0.25
barbatus Andr.
Lamiaceae EPH 446
173 Plumbago zeylanica Vellaikoduvilai He Lf/Rt Paste Topical Pb 5 (Snake bite) 0.92 0.42
L. SMSD 6 (Arthritis)
Plumbaginaceae
EPH 630
174 Polygala javana Palapirantai He Lf Paste Topical GUA 6 (Breast pain) 0.50 0.17
DC. var. angustifolia
Polygalaceae EPH
631
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
83
Table 2 (continued)
84
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
175 Pongamia pinnata Pongan Tr Sd Raw Topical RSD 6 (Cold & cough) 0.75 0.58
(L.) Pierre (Necklace) LP 3 (Jaundice)
Fabaceae EPH 160
176 Portulaca pilosa L. Pillaiva- He Tu Raw Oral GUA 3 (Prevent natural 0.25 0.67
subsp. pilosa larthikilangu abortion)
Portulacaceae EPH
161
177 Portulaca wightiana Pasalai chedi He Wp Paste Topical SMSD 5 (Arthritis) 0.42 0.58
Wall.ex Wight &
Arn.
Portulacaceae EPH
162
178 Pouzolzia zeylanica Kallazhingi He Rb Paste Topical He 6 (Dandruff) 1.17 0.67
(L.) Benn. Pi Raw/paste Topical SMSD 8 (Swelling,
Urticaceae EPH 155 muscular pain, bone
fracture)
179 Premna corymbosa Aadhandai Sh Lf Raw (crushed) Topical SMSD 3 (Giddiness) 0.25 0.67
(Burm, f.) Rottl. & (inhalation)
Willd.
Verbenaceae EPH
629
180 Priva cordifolia (L. Aadai otti He Lf Paste Topical DID 7 (Wounds) 0.58 0.17
f.) Druce.
Verbenaceae EPH
469
181 Pterocarpus Vengai Tr Re Soaked Topical SMSD 9 (Joint pain, 2.25 0.83
marsupium Roxb. Lf Paste Topical rickets)
Fabaceae EPH 470 St Decoction Oral DID 7 (Leucoderma, skin
diseases)
P. S. Tresina et al.
ED 11 (Diabetes)
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
182 Pupalia lappacea Pakkattikai He Ts/If Paste Topical Pb 2 (Unknown insect bite) 0.17 0.25
(L.) Juss. var.
lappacea
Amaranthaceae
EPH 619
183 Rhinacanthus Nagamalli Sh Lf Juice/ Oral Pb 10 (Snake bite) 0.83 0.83
nasutus (L.) Kurz powder
var. nasutus
Acanthaceae EPH
526
184 Rubia cordifolia L. Koduvili He Rt/Ts Paste Topical DID 3 (Weapon injury) 0.25 0.25
Rubiaceae EPH 360
185 Sansevieria Maurl He Lf Raw Topical (bite) DC 3 (Teeth growth in 0.25 0.33
roxburghiana young children)
Schultes & Schultes
Agavaceae EPH 98
186 Santalum album L. Sandanam Tr Hw Paste Topical CA 12 (Body cooling) 1.00 0.58
Santalaceae EPH 99
187 Scoparia dulcis L. Neernangai He Lf Raw Oral Pb 8 (Poison bite) 2.42 0.83
Scrophulariaceae Wp Decoction ED 12 (Diabetes)
EPH 120 GIA 9 (Stomachache)
188 Senna auriculata Aavarai Sh Fl Paste/juice Oral GIA 8 (Colic pain) 3.00 0.91
(L.) Roxb. Lf Ts Juice Topical (tied) ED 12 (Diabetes)
Caesalpiniaceae Raw GIA 10 (Laxative)
EPH 50 GIA 6 (Colic pain)
189 Sesamum indicum Kattuyellu He Sd Raw Oral GUA 8 (Abortion) 0.67 0.67
L.
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
Pedaliaceae EPH 51
(continued)
85
Table 2 (continued)
86
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
190 Sesbenia sesban (L.) Chittakatti Sh Lf Boiled (coconut Topical HC 9 (Graying Greying of 1.50 0.50
Merr. oil) Oral hair, hair growth)
Fabaceae EPH 52 Juice GIA 9 (Bile complaints)
191 Sida cordata Nilathuti He Wp/Rt Decoction Oral RSD 10 (Asthma) 2.17 0.67
(Burm.f) Borssum Fl Paste Oral GUA 8 (Diuretic, urinary
Malvaceae EPH 660 disorder) GIA 8 (Ulcer,
piles)
192 Sida rhombifolia L. Karunthatti He Rt Decoction Oral Fvr 12 (Fever) 1.00 0.33
var. rhombifolia
Malvaceae EPH 661
193 Solanum anguivi Kattuthuduvalai He Uft Roasted (Curry) Oral RSD 7 (Dry cough) 0.58 0.50
Lam.
Solanaceae EPH
180
194 Solanum nigrum L. Thaka keerai He Lf Cooked Oral DID 7 (Somatitis) 1.92 0.67
Solanaceae EPH Uft (curry) Oral RSD 6 (Wheezing)
181 Cooked GIA 10 (Stomachache,
ulcer)
195 Solanum surattense Kandangathri He Lf Decoction Oral RSD 12 (Cold and cough) 1.00 0.83
Bunn. f.
Solanaceae EPH
182
196 Solanum torvum Sw Sundaikkai Sb Ft Raw/cooked Oral LP 6 (Jaundice) 2.5 0.83
Solanaceae EPH GIA 9 (Ulcer,
183 anthelmintic)
RSD 12 (Cold and cough)
197 Solanum trilobatum Thuduvalai Cl Lf Raw (roasted)/ Oral RSD 12 (Cold and cough) 1.00 0.83
L. Uft juice
P. S. Tresina et al.
Solanaceae EPH
184
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
198 Sphaeranthus Oothuchedi He Lf Raw (squeezed) Topical SMSD 6 (Giddiness) 0.50 0.50
indicus L (smelled)
Asteraceae EPH 620
199 Sterculia urens. Vennaali Tr Es Roasted Oral GUA 6 (Lactation) 0.50 0.17
Roxb
Sterculiaceae EPH
598
200 Strychnos Kanjirai Tr St Raw Topical (kept GH 3 (Ill effect of evil 0.75 0.25
nux - vomica L. Sd Ash inside the spirit)
Loganiaceae EPH house) SMSD 6 (Rickets/Rgchitis)
333 Topical
201 Swertia angustifolia Milagainagai He Wp Powder Oral Pb 4 (Snake bite) 0.83 0.42
Buch. – Ham var. GIA 6 (Stomachache)
pulchella (Buch-
Ham ex D.Don)
Burkill
Gentianaceae EPH
234
202 Swertia corymbosa Milagainagai He Wp Powder Oral Pb 3 (Snake bite) 0.75 0.33
(Griseb.) Wight ex GIA 6 (Stomachache)
Clarke var.
corymbosa
Gentianaceae EPH
235
203 Symplocos Tulsimanimarm Tr Sd Raw Topical SMSD 6 (Rickets/Rgchitis) 0.50 0.33
cochinchinensis (Necklace)
(Lour.) Moore
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
subsp. laurina
(Retz) Nooteb
Symplocaceae EPH
87
421
(continued)
Table 2 (continued)
88
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
204 Syzygium cumini Naval Tr Sd Powder Oral ED 12 (Diabetes) 1.00 0.91
(L.) Skeels
Myrtaceae EPH 422
205 Tabernaemontana Nandhiavattai Sh Pt Juice Topical ENT 10 (Improve vision) 0.83 0.67
heyneana Wall.
Apocynaceae EPH
115
206 Tamarindus indica Puli Tr Ft Raw Oral CSCD 8 (Cholesterol 1.58 0.67
L. Sd Raw Topical lowering)
Caesalpiniaceae Sb Decoction (Rubbed) Pb 5 (Scorpion sting)
EPH 23 Oral ENT 6 (Sore throat)
207 Tephrosia purpurea Kolingi He Rb Juice Oral GIA 7 (Bowel disorder) 0.58 0.17
(L) Pers.
Fabaceae EPH 116
208 Tephrosia villosa Kuruttukolingi He Lf Paste Topical DID 6 (Eczema) 0.50 0.17
(L.) Pers.
Fabaceae EPH 117
209 Terminalia bellirica Tani Tr Pc Decoction Oral Fvr 7 (Fever) 2.33 0.67
(Gaertn.) Roxb. Ft Raw RSD 7 (Asthma,
Combretaceae EPH Sb Decoction bronchitis)
613 GIA 6 (Anthelmintic)
GUA 7 (Diuretic)
210 Terminalia chebula Kadukai Tr Pc Paste Oral GIA 7 (Dysentry) 2.58 0.67
Retz. Sb Decoction GUA 7 (Diuretic)
Combretaceae EPH Ft Powder CSCD 5 (Cardio tonic)
614 GIA 12 (Laxative)
211 Thespesia populnea Puvarasu Tr Ts Paste Topical Pb 5 (Scropion sting) 0.42 0.33
(L.) Soland.ex
P. S. Tresina et al.
Correa
Malvaceae EPH 621
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
212 Tinospora cordifolia Enthalkodi Cl St Infusion/ Oral LP 6 (Jaundice) 3.16 0.91
(Willd) Miers ex St decoction Oral GIA 6 (Anthelmintic)
Hook. f & Thomas Rt Juice Oral Fvr 10 (Fever)
Menispermaceae Wp Juice Pb 6 (Antidote, snake bite,
EPH 622 scorpion sting)
SMSD 10 (Rheumatism)
213 Toddalia asiatica Milagaranai Sh Sb Powder Oral SMSD 10 (Rheumatism) 0.83 0.58
(L.) Lam var.
floribunda
Rutaceae EPH 678
214 Toddalia asiatica Milagaranai Sh Ft Juice Oral GIA 8 (Improve digestion) 0.67 0.50
(L.) Lam var.
gracilis
Rutaceae EPH 679
215 Tribulus terrestris L. Sirunerinji He Ft Decoction Oral GUA 12 (Diuretic, urinary 1.67 0.67
Zygophyllaceae Wp Juice Oral disease, antiurolithic
EPH 24 aphrodisiac)
GIA 8 (Anthelmintic)
216 Trichodesma Kavuthumbai He Lf Roasted Oral GIA 7 (Piles) 0.58 0.67
zeylanicum (Burm.
f.) R. Br.
Boraginaceae EPH
662
217 Tridax procumbens Mukoothielai He Lf Juice Topical SMSD 6 (Shoulder pain) 1.50 0.67
L Paste DID 12 (Eczema, wounds,
Asteraceae EPH 15 weapon injury)
218 Tylophora indica Nanchianuppan Cl Lf Juice Oral GUA 6 (Diuretic) 2.17 0.58
(Burm. F.) Merr palai Powder RSD 10 (Asthma,
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled…
(continued)
Table 2 (continued)
90
Botanical name,
family, voucher Vernacular Part Ailment category: no. of
S. no. specimen no. name Life form used Preparation Application use-reports Use value RFC
219 Vetiveria zizanioides Vetiver He Rt Decoction Topical DID 10 (Skin diseases) 1.67 0.58
(L.) Nash HC 10 (Hair tonic, hair
Poaceae EPH 21 loss)
220 Vitex altissima L. f. Mayila- Tr Sb Boiled Topical (bath) ENT 3 (Burning sensation 0.25 0.17
Verbenaceae EPH dunkuringi in the eyes)
664
221 Vitex negundo L. Vennochi Sh Lf Roasted Topical SMSD 4 (Headache) 2.00 0.75
Verbenaceae Wp Decoction (foment) GUA 3 (Diuretic)
EPH 22 Oral GIB 7 (Anthelmintic)
RSD 10 (Expectorant)
222 Waltheria indica L. Thuverpuli He Rb Infusion Oral GIA 5 (Bowel disorder) 0.42 0.25
Sterculiaceae EPH
602
223 Wrightia tinctoria Vetpalai Tr Lf Soaked Topical DID 5 (Eczema) 0.42 0.33
(Roxb.) R.Br. var. (coconut oil)
tinctoria
Apocynaceae EPH
534
224 Ziziphus xylopyrus Mullukottai Tr Sb Infusion Oral GIA 6 (Stomachache) 0.50 0.42
(Retz.) Willd
Rhamnaceae EPH
702
Part used: Lf Leaf, St Stem, Sb Stem bark, Rt Root, Rb Root bark, Ap Aerial part, Wp Whole plant, Ts Tender shoot, Yp Young plant, Bb Bulb, Hw Heart wood,
Ft Fruit, Uft Unripe fruit, If Inflorescence, Fl flower, Sd Seed, Isd Immature seed, Rc Rachis, Re Resin, Pc Pericarp, Es Endosperm, Yes Young endosperm, Sl
Stem latex, Lx Latex, Ss Stem sap, Rh Rhizome, Tu Tuber
Life form: He Herb, Sh shrub, Tr Tree, Cl Climber, Wcl Woody climber, CHe Climbing herb, She Succulent herb, Su Succulent
P. S. Tresina et al.
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 91
Series1, Climber,
18%, 18%
Series1, Tree, 23%,
23%
16, 18] [42–46]. Leaves are dominantly used in making herbal preparations due to
the presence of bioactive constituents. Leaves were preferably used by the local
people because leaves are easily removed as compared to whole plant and roots that
also damage the growth of plants, which leads to the decline in population of medic-
inal plants [47, 48].
92 P. S. Tresina et al.
Tender shoot, 2%
The preparation and utilization of plant parts used were grouped into nine categories
(Fig. 8). Among these, majority of the plant remedies were prepared by paste (35%)
followed by juice (24%), raw (24%), powder (16%), roasted (13%), decoction
(12%), boiled (8%) infusion (7%) and cooked (5%). Preparation of paste for the
treatment of ailments is a common practice among the other tribal people in global
level [8, 18, 49–53]. The paste was prepared by grinding the fresh or dried plant
parts in the water or oil. The decoction was obtained by boiling the plant parts in
water until the volume of the water is reduced to minimum or the required quantity.
Infusion was prepared by soaking fresh plant parts in water over night. The inhala-
tion was done by the burning of plant parts and the smoke is inhaled through nose.
Most of the informants suggested taking herbal medicines orally (74%), rather
than external (55%) use, in consistent with comparable investigations [54, 55]. For
topical use, the most important methods used were direct application of paste or
medicated oil and mostly dealt with diseases such as skin disorders, wounds, heel
crackers, poison bite, rheumatism, body pain, headache and hair growth. Most of
the medicines were given orally which is in agreement with some other studies
conducted elsewhere [8, 12, 17, 56, 57]
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 93
The medicinal arrangements were completed out of a single plant part or in mixture
of many plant parts. In case of Paliyar’s medicinal preparation, single mode (184
plants) of preparation was dominating over the multiple modes (40 plants) of prepa-
ration. In some cases, Paliyar traditional healers too frequently use some adjuvants
such as honey, cow/goat milk, coconut oil, salt, gingery oil and palm jagging
(Table 3). Paliyar traditional healers used in the multiple mode preparation, more
than two or more plant parts for the preparation of medicine in the treatment of
single or multiple ailments. Similar findings were reported by some researchers
[16–18]. The use of multiple plant remedies among the traditional healers could be
attributed to the belief of synergic reactions where one plant could have a potentiat-
ing effect than other [50]. In this study, mostly fresh plant parts were used for the
preparation of medicine. Similar findings were reported by Assae et al. [58]; Revathi
et al. [59]; Yabesh et al. [18] and Xavier et al. [17]. Local traditional healers too
frequently used other adjuvant like honey, milk, sugar, salt and oil to improve the
acceptability and medicinal property of certain remedies. The preparation of paste/
medicated oil was commonly used by the oil of coconut and gingery oil. They were
using specific plant parts and specific dosages for the treatment of diseases and the
dose given to the patient depends on their age, physical status and health conditions.
Before giving treatment, the condition of the patients was observed deeply and then
they were given the prepared medium.
94 P. S. Tresina et al.
Table 3 Ingredients added for the preparation of herbal medicines by the Paliyar traditional healers
Other plants added in medicinal
Botanical name preparation Other ingredients added
A. columnaris Terminalia chebula Honey
A. indica – Calcium hydroxide
A. aspera – Lime
A. scholaris – Milk, rice fermented
water
A. sylvaticus Tamarindus indica Coconut oil
A. subdecurvens – Black soil
A. tagala – Urine of a child of
opposite sex
A. racemosus Curcuma longa –
B. pilosa var. minor Phyllanthus amarus Milk
B. javanica Dodonaea viscosa Coconut oil
C. separia – Lemon juice
C. perfoliata – Rice fermented water
C. halicacabum Oriza sativa, Salt
Cuminum cyminum, Solanum
trilobatum,
S. surattense, Ferula asafoetida
C. asiatica – Milk
C. quadrangularis Ferula asafoetida –
C. hirsutus – Milk
C. adansonii – Coconut oil
C. dactylon Cumimum cyminum –
C. cinerea – Milk, rice fermented
water
D. palmatus – Ghee
D. viscosa Bischofia javanica Coconut oil
E. discifera – Milk, rice fermented
water
E. alsinoides Ocimum tenuiflorum, Sida –
rhombifolia var. rhombifolia
F. indica – Milk
G. resinifera – Milk (dry cough)
G. rottleriformis – Milk
G. marantina Plectranthus barbatus –
G. superba – Coconut oil
H. isora – Coconut oil
H. indicus var. indicus – Milk
H. indicus var. pubescens – Milk
H. rigens var. rigens – Salt
H. vitifolius Allium cepa –
H. integrifolia Allium cepa –
(continued)
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 95
Table 3 (continued)
Other plants added in medicinal
Botanical name preparation Other ingredients added
I. obscura Allium cepa –
J. roxbhurghianum Curcuma longa (colic pain) Gingelly oil (oozing pus
from the ear)
K. grandiflora – Coconut oil, black soil
L. camara var. aculeata Allium cepa, Mimosa pudica –
L. inermis – Coconut oil
L. nicotianifolia var. Piper betle, Areca catechu –
nicotianifolia
M. indica Curcuma longa –
M. biflora – Coconut oil
M. pudica Lantana camara var. aculeata, –
Allium cepa
M. elengi – Rice fermented water or
milk
M. cerviana Phyllanthus emblica, Terminalia Palm jaggery
bellirica, T. chebula, Zingiber
officinale, Alpinia calcarata, Piper
longum, Ocimum tenuiflorum,
Mukia maderaspatana, Evolvulus
alsinoides
M. pentaphylla Curcuma longa, Plectranthus –
barbatus
M. pubescens var. pubescens Allium cepa, curcuma longa Coconut oil
M. concanensis Piper nigrum, Allium sativum –
M. atropurpurea Tamarindus indica –
M. maderaspatana – Calcium hydroxide
M. koenigii Cocos nucifera –
N. colebrookeana – Milk
P. daemia Allium sativam –
Erythrina variegata, Papaver
somniferum
P. amarus – Milk
P. maderaspatensis – Milk
P. minima Ferula asafoetida, Allium cepa Coconut oil, salt
P. argyrophyllum Trachyspermum ammi, Piper –
longum, Zingiber officinale
P. trioicum Vitex altissima Coconut oil
R. nasutus var. nasutus – Milk
S. auriculata Ferula asafoetida, Papaver –
somniferum
S. indicum – Palm jaggery
S. sesban – Coconut oil
S. rhombifolia var. rhombifolia Evolvulus alsinoides, Ocimum –
tenuiflorum
(continued)
96 P. S. Tresina et al.
Table 3 (continued)
Other plants added in medicinal
Botanical name preparation Other ingredients added
S. anguivi var. anguivi – Coconut oil
S. surattense Solanum trilobatum, Cuminum –
cyminum, Ferula asafoetida
S. trilobatum Coriandrum sativum Ghee, salt
S. urens – Ghee
S. nux - vomica – Castor oil
Coconut oil
S. angustifolia var. pulchella – Urine of a child of
opposite sex
S. corymbosa var. corymbosa – Urine of a child of
opposite sex
T. purpurea Ferula asafoetida
T. bellirica Phyllanthus emblica, Terminalia Palm jaggery
chebula, Zingiber officinale,
Alpinia calcarata, Piper longum,
Ocimum tenuiflorum, Mollugo
cerviana, Mukia maderaspatana,
Evolvulus alsinoides
T. chebula Curcuma longa –
T. populnea Allium cepa –
T. zeylanicum Allium cepa –
T. procumbens – Calcium hydroxide
V. altissima Piper trioicum Coconut oil
V. zizanioides Hemidesmus indicus var. indicus, Coconut oil
Hedyotis puberula
W. tinctoria var. tinctoria – Coconut oil
The most commonly used species was Tinospora cordifolia with 38 use reports by
12 informants, giving the highest use value of 3.16. T. cordifoliais attributed to its
use in the treatment of various diseases, and it is well recognized by all informants
as the plant having medicinal value. T. cordifoliais is known for its immense appli-
cation in the treatment of various diseases in the traditional Ayurvedic literature.
Recently, the discovery of active components from the plant and their biological
function in disease control have activated interest across the globe [60].
Other important plants with high use value were Cynodon dactylon (37 use
reports by 12 informants with a UV value of 3.08) followed by Senna auriculata (36
use reports by 12 informants with UV of 3.00), Hemidesmus indicus var. pubescens
(35 use reports by 12 informants with a UV of 2.92), H. indicus var. indicus (34 was
reported by 12 informant with UV of 2.83), Terminalia chebula (31 use reports by
12 informants with a UV of 2.58), Andrographis paniculata, Azadirachta indica,
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 97
To gain credibility, scientific studies that use traditional knowledge must be depend-
able. In ethnobotanical studies, consensus analysis provides a measure of reliability
for any given claim providing reliable evidence. General Fic of local knowledge for
disease treatment depended on the availability of the plant species in the study area
[51]. In order to use the informant consumer factor (Fic), the researchers clarified
the illnesses into broad categories (16 categories).
The Fic values in the present study ranged from 0.83 to 1.00 (Table 4). The cat-
egories with more than 200 use reports were gastrointestinal ailments (541 use
reports, 65 species) skeleto-muscular system disorders (451 use reports, 56 spe-
cies), dermatological infections/diseases (317 use reports, 48 species), respiratory
system diseases (278 use reports, 31 species) and genitor-urinary ailments (208 use
reports, 29 species). In the present study, oncology, hair care, circulatory system/
cardiovascular disease, cooling agents, endocrine disorder and fever had the highest
98 P. S. Tresina et al.
Fic of 1.00, 0.91 and each 0.90, respectively. In this study, oncology had the highest
Fic of 1.00, and it is in the agreement with the previous studies among the neigh-
bouring indigenous communities in Kerala. India [18]. In the present study, hair
disease and endocrinal disorders also had the highest Fic of 0.91 and 0.90, respec-
tively, whereas hair disease and endocrinal disease had the highest Fic of 1.00
among the Kani tribes in Thoduhills of Kerala [17] and Irulas in the Tanjore district
[62]. Syzygium cumini, Gymnema sylvestris and Momordica charantia var. chara-
nita had been very commonly used for the treatment of diabetes, and Micromeria
biflora, Eclipta prostrata and Pouzolzia zeylanica were used for the treatment of
hair problems.
The least agreement between the informants was observed in poisonous bite with
a Fic of 0.83 followed by dermatological infections/diseases with a Fic of 0.85; ear,
nose, throat and eye problems; general health and genitor-urinary ailment with a Fic
of 0.86, respectively. Thus, the study indicates that the degree of knowledge shared
by the users in the study is regarding the use of medicinal plants in the treatment of
ailments is high. Dermatological infections/diseases had the lowest Fic of 0.85, but
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 99
this ailment category ranked third in the number of use reports 317 and of taxa 48
attributed to this category. It may be due to the lack of communication among the
informants in the study area who are involved in the ailment group [61] or it may be
due to the lack of dermatological disorders among the studied tribal people [17].
The analysed categories with major agreements from the higlight and the most
important plants in each category are shown in Table 5. Of the reported plants, 39
species had the highest fidelity level of 100% most of which were used in the single
ailment category with multiple informants. The plants with the highest FL of 100%
were Ficus microcarpa (DID), Gloriosa superba, Tabernaemontana heyneana,
Commelina benghalensis, C. ensifolia (ENT), Syzygium cumini, Memecylon edule,
Eleusine coracana (ED), Sida rhombifolia var. rhombifolia, Limonia acidissima,
Lantana wightiana (Fvr), Amorphophallus sylvaticus, Indigofera wightii, Physalis
minima, Lepisanthes senegalensis (GIA), Caralluma adscendens var. attenauta,
C. lasiantha (GH), Cleome viscosa, Curculigo orchioides, Diplocyclos palmatus,
Ensete superbum, Hybanthus enneaspermus, Mimusopis elengi (GUA), Micromeria
bifolra, Eclipta prostrata (HC), Bidens pilosa var. minor, Phyllanthus amarus,
P. maderaspatensis (LP) Rhinacanthus nasutus var. nasutus, Justicia glauca (Pb)
Justicia adhatoda, Piper argyrophyllum, Solanum trilobatum, S. surattense (RSD),
Acorus calamus, Anisomeles malabarica, A. indica, Capparis zeylanica and
Mucuna atropurpurea (SMSD).
The maximum FL for the above plants indicated the 100% choice of the inter-
viewed informants for treating specific ailments, and this could be an indication of
their healing potential. In support to our study, 100% FL was reported in P. amarus
for jaundice among the herbal healers in Shimoga district of Karnataka [51], Malasar
tribes in Velliangarihills of Tamil Nadu [63], Kani tribes of Tirunelveli hills of
Western Ghats, Tamil Nadu, India [16] and Kani tribes in Thodu hills of Kerala,
South India [17].
The RFC shows the local importance of every species with reference to the infor-
mants who cited uses of these plant species [39]. In the present study, it ranged from
0.91 to 0.17 (Table 2). Achyranthes aspera, Acorus calamus, Aegle marmelos,
Allium cepa, Aloe vera, Andrographis paniculata, Azadirachta indica, Calotropis
gigantea, Caralluma adscendens var. attenauta, C. lasiantha, Commelina bengha-
lensis, C. ensifolia, Coriandrum sativum, Curculigo orchioides, Cymbopogon citra-
tus, Cynodon dactylon, Eclipta prostrata, Eleusine coracana, Ensete superbum,
Gymnema sylvestre, Hemidesmus indicus var. indicus, H. indicus var. pubescens,
Table 5 Fidelity level (FL) values for common medicinal plants used by Paliyar traditional healers by ailment categories
100
4 Conclusion
The present study revealed that traditional medicines were still in common use by
the Paliyar tribal communities and accurate knowledge of the plants and their
medicinal properties were held by only a few individuals in this community. Hence,
a need for detailed investigation of ethnobotanical knowledge held by each tribal
community is required before such valuable knowledge vanishes. Thus, the present
An Ethnobotanical Study of Medicinal Plants Used by Traditional Healers in Grizzled… 103
study would be useful in preventing the loss of ethno medicinal traditions of Paliyar
tribal Communities. The plants with high fidelity level, use values and relative fre-
quency citation in the present study may indicate the possible occurrence of valu-
able phytochemical compounds, and it requires a search for probable new drugs to
cure different ailments. The efficacy and safety of all the reported ethnomedicinal
plants need to be evaluated for phytochemical and pharmacological studies espe-
cially the plants with high informant consensus factor, use value, fidelity level and
relative frequency citation should be given priority to carry out bioassay and toxic-
ity studies. The present study suggested that the plants having higher use reports
such as Tinospora cordifolia, Cynodon dactylon, Senna auriculata, Hemidesmus
indicus var. indicus, H. indicus var. pubescens, Terminalia chebula and Andrographis
paniculata need further investigation in regard to phytochemical and associated
pharmacological studies.
Acknowledgement The authors express their thanks to Paliyar tribes in the study area for reveal-
ing their traditional knowledge.
Conflict of Interest No conflicts of interest were reported by the authors.
References
51. Rajakumar N, Shivanna MB (2009) Ethno-medicinal application of plants in the eastern region
of Shimoga District, Karnataka, India. India J Ethnopharmacol 126:64–73
52. Ullah M, Usman Khan M, Mahmood A, Hussain M, Mehmood Wazir S (2013) An ethnobotan-
ical survey of indigenous medicinal plants in Wana district South Waziristanagency, Pakistan.
J Ethnopharmacol 150:918–924
53. Hussain S, Hussain W, Nawar A, Badshah L, Ali A, Ullah S et al (2022) Quantitative ethno-
medicinal study of indigenous knowledge on medicinal plants used by the tribal communi-
ties of central Kurram, Khyber, Pakhtunkhwa, Pakistan. Ethnobot Res App 23:5. https://doi.
org/10.32859/era23.5.1-31
54. Kayani S, Ahmad M, Sultana S, Khan Shinwari Z, Zafar M, Yaseen G et al (2015) Ethnobotany
of medicinal plants among the communities of Alpine and Sub-alpine regions of Pakistan. J
Ethnopharmacol 164:186–202
55. Sadat-Hosseini M, Farajpour M, Boroomand N, Solaimani-Sardou F (2017)
Ethnopharmacological studies of indigenes medicinal plants in the South of Kerman, Iran. J
Ethnopharmacol 199:194–204
56. Ribeiro A, Romeiras MM, Tavares J, Faria MT (2010) Ethnobotanical survey in Canhane
village, district of Massingir, Mozambique: medicinal plants and traditional knowledge. J
Ethnobiol Ethnomed 6:33. http://www.ethnobiomed.com/content161133
57. Khan I, Abd Elsalam NM, Fouad H, Tariq A, Ullah R et al (2014) Application of ethnobotani-
cal indices on the use of traditional medicines against common diseases. Evi based Comple
Alter Med 2014:1. https://doi.org/10.1155/2014/635371
58. Assae A, Akwetey GA, Achel DA (2010) Ethnopharmacological use of herbal remedies for the
treatment of malaria in the Dangme West District of Chinna. J Ethnopharmacol 129:367–376
59. Revathi P, Parimelazhagan T, Manian S (2013) Ethnomedicinal plants and novel formulations
used by Hooralis tribe in Sathyamangalam forests, Western Ghats of Tamil Nadu, India. J Med
Plants Res 7:2083–2097
60. Saha S, Ghosh S (2012) Tinospora cordifolia: one plant, many roles. Anc Sci Life 31:151–159
61. Rokaya MB, Munzbergova Z, Timsina B (2010) Ethnobotanical study of medicinal plants
from the Humla district of western Nepal. J Ethnopharmacol 130:485–504
62. Regupathy S, Newmaster S (2009) Valorizing the ‘Irulas’ traditional knowledge of medicinal
plants in the Kodiakkarai Reserve Forest, India. J Ethnobiol Ethnomed 5:10
63. Regupathy S, Steven NG, Maruthakkutti M, Velusamy B, Ul Huda MM (2008) Consensus of
the Malasars traditional aboriginal knowledge of medicinal plants in the Velliangiri holy hills,
India. J Ethnobiol Ethnomed 4:8
64. Mukherjee PK, Nema NK, Venkatesh P, Debnath PK (2012) Changing scenario for promotion
and development of Ayurveda-way forward. J Ethnopharmacol 103:25–35
65. Amjad MS, Qaeem MF, Ahmed I, Khan SU, Chaudhari SK et al (2017) Descriptive study of
plant resources in the context of the ethnomedicinal relevance of indigenous flora: a case study
from Toli Peer National Park, Azad Jammu and Kashmir, Pakistan. PLoS One 12. https://doi.
org/10.1371/journal.pone.0171896
66. Camou-Guerrero A, Reyes-Garcia V, Martinez-Ramos M, Casas A (2008) Knowledge and
use value of plant species in a Raramuri community: a gender perspective for conservation.
Human Ecol 36:259–272
Ethnomedicinal Plants Used by Irula
Tribal Settlement of Attappady
in Palakkad District, Kerala, India
1 Introduction
Biologically active compounds present in plants account for their use in traditional
medicinal systems for centuries. 80% of the world’s population depends on plants
for their healthcare (World Health Organization,1993). Traditional communities are
intimately familiar with the use of medicinal plants, which is unknown the modern
society [1, 2]. This practice of health care is based on the belief and experience of the
traditional people, which is a part of their tradition and culture. The modern health
care system relies mainly on plant-based ingredients. The traditional use of wild
plants not only provides folk medicines for the health care system but also as a food
source for local communities. The ethnic people practiced indigenous system of folk
medicines for centuries and their knowledge about the usage and maintenance of
plants was passed from generation to generation especially through mouth [3–5].
Kerala is lying in the southern end of the Indian subcontinent. The state is known
for its cultural and biological diversity. As per the 2011 Census, the tribal popula-
tion in Kerala is 364,189, which is 1.14% of the total population of the state. The
decadal growth of tribal population in the state has been 13.75% [6, 7].
Attappadi is part of three states-Kerala, Tamil Nadu and Karnataka. It is located
in the mid-eastern part of Kerala. It is a segment of Western Ghats ranges. Attappadi
can be considered as an extension mountain valley, which has an area of 731 sq.km.
Attappadi is a part of Palakkad district, adjoining Coimbatore and Nilgiri districts of
Tamil Nadu. The population of Attappadi consists of both tribes and non-tribes [8, 9].
The original version of this chapter was revised. The correction to this chapter is available at
https://doi.org/10.1007/978-3-031-28780-0_65
The name Attappadi means atta, the blood leach and pad, the habitation. There
are three different major tribal communities in Attappadi, they are Irulars, Mudugars
and Kurumbas. Tribal groups in Attappadi are the most backward than other tribal
communities of Kerala. Their culture is traditional in nature and they depend mainly
on land and forest for their livelihood [9].
Tribal settlements in Attappadi are known as Ooru (hamlet). There are 187 ham-
lets in Attappadi. The Irulas were inhabiting the plains and low elevations of
Attappadi constituted the majority tribal population [9, 10]. The Attappadi Block is
divided into the three Gram Panchayats—Agali, Pudur and Sholayur. The number
of Irula oorus is 53, 43 and 44 in Agali, Pudur and Sholayur, respectively [8].
Traditional knowledge of human health and medicine has recently become a
universal consideration. The value of medicinal plants and herbs is being lost due to
lack of awareness and deforestation. The ethnobotanical study is the collection of
ethnobotanical information and documentation of traditional knowledge. It has
gained importance as it saves knowledge that is transferred from generation to gen-
eration only inside a particular community. The present study aims to document the
plants used by Irula tribes of Palakkad district of Kerala state.
2.1 Study Area
The study was conducted in Attappadi, one of the tribal areas of Kerala, which is
part of the Western Ghats of India. The valley is an extensive east sloping plateau on
the Western Ghats of Kerala that covers an area of 745 km2, which is located in the
Mannarkad taluk of Palakkad district. Geographic coordinates of Attappadi are 3“N
and 76°33’43.4”E [9, 11].
Attappadi is a part of the Nilgiri Biosphere Reserve. The population of Attappadi
consists of adivasis and non-adivasis. Irulas, Mudugas and Kurumbas are three
tribal groups inhabiting the region of which, Irulas inhabiting the plains and low
elevations, constitute the majority (79%). There are 187 hamlets in Attappadi, which
are inhabited by both the adivasis and the non- adivasis. Each Ooru contains, on
average, 50 houses. The tribal people in attappadi were most backward among the
tribal groups of Kerala and their livelihood depends mainly on land and forest. The
Attappadi comprises three Gram Panchayats – Agali, Sholayur and Pudur [8, 12].
2.2 Data Collection
The data collection was done through field surveys conducted at the study area. It
was carried out by interviewing traditional healers in different locations of Irula
tribal settlement in a period between February to July 2020. Questionnaires were
structured for the interview and were applied to 29 informants (12 men and 17
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 109
women) who are in age between 30 and 70 years to gather information about plants
that are used by the tribal people for various purposes. The informants were selected
based on their popularity among other local people with respect to the knowledge
about medicinal plants.
Data such as local names, uses for various ailments and disease were collected
from traditional healers and other tribes through individual interviews during the
survey. The plants were also documented by using digital photography, and voucher
specimens were collected along with the survey and the collected specimens were
identified using different floras. Voucher specimens of this study have been depos-
ited at the Herbarium of Department of Botany, Government Victoria College,
Palakkad. The botanical names of the plant specimens were updated according to
the Plant List.
The ethnobotanical information collected was analysed for obtaining the impor-
tance of individual species. The data obtained were classified into relatively well-
defined ethnomedical categories to get a clear picture about the use of particular
plant species.
Informant Consensus Factor (Fic)
The informant consensus factor (Fic) was used to see if there was agreement in the
use of plants in the ailment categories between the plant users in the study area. The
Fic was calculated.
by the following formula:
Nur Nt
Fic
Nur 1
Fic, which is the ratio of the number of use reports in each category (Nur) minus
the number of taxa used for a particular ailment category by all informants (Nt) and
the number of use reports in each category minus one. Thus, Fic value close to 1 was
taken as an indication of high intracultural consensus, i.e. more healers use the same
species, whereas a value close to zero was regarded as a low probability of medici-
nal use [13].
The present study aimed to document the plants used by the Irula tribal settlement
of Attappady, Palakkad district. Information regarding the use of medicinal plants
was collected from 29 informants, 12 males and 17 females aged about 30–70 years.
Informants were traditional healers and local people. From the study,
110 C. V. Jayalekshmi et al.
ethnobotanical information of 126 plant species was recorded. The most cited fami-
lies were Leguminosae, Apocynaceae, Amaranthaceae, Euphorbiaceae and
Lamiaceae. The photographs of some plants cited in the study are given in Fig. 1.
The Irula tribes use a lot of plants as food, medicine, fodder and for making mats,
hut, etc. Some plants are grown as sacred plants, such as Aerva lanata, Azadirachta
indica, Cynodon dactylon, Ficus religiosa and Ocimum sanctum.
The information on plants used by Irula tribes is given in Table 1. Scientific name,
family, common name and medicinal importance of plants are documented in Table 1.
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
22 Bambusa bambos Poaceae Mungil Anti-inflammatory,
(L.) Voss febrifuge
23 Basella alba L. Basellaceae Vasala Fever, stimulant
and expulsion of
worms, used to
treat back pain
24 Basella Basellaceae Sivpuvasali Useful to diuretic,
paniculata gonorrhoea
Volkens
25 Bauhinia Leguminosae Ashamaram. Bark and fibre Veterinary
racemosa Lam. medicine
26 Bauhinia Leguminosae Mantharia Cough, stomach
variegata L. problem
27 Boerhavia Nyctaginaceae Cerandhia Stomach ulcer,
diffusa L. antibacterial,
diuretic,
anti-helminthic
28 Brassica juncea Brassicaceae Kaduku Treat inflammation
(L.) Czern. Treat abdominal
pain
29 Cajanus cajan Leguminosae Thuvarai Jaundice, oral
(L.) Millsp. problem
30 Calotropis Apocynaceae Losandi Anti-inflammatory
gigantea (L.)
Dryand.
31 Capsicum annum Solanaceae Mulaku Treatment of ulcer,
L. cancer,
anti-inflammatory
32 Cardiospermum Sapindaceae Stomach upset,
halicacabum L. headache
33 Carica papaya Caricaceae Pappaly Stomach ache,
L. digestive problem
34 Cascabela Apocynaceae Arali maram Controls back pain
thevetia (L.)
Lippold
35 Cassia fistula L. Leguminosae Kanikundri Febrifuge,
astringent, used
against ringworm,
jaundice
36 Cassia tora L. Leguminosae Thira Stomach pain, Veterinary
medicine
37 Catharanthus Apocynaceae Nithyakalyani Anti-neoplastic,
roseus (L.) sedative
G. Don
(continued)
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 113
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
38 Celastrus Celastraceae Laxative, digestive
paniculatus problems, skin
Willd. disease
39 Centella asiatica Apiaceae Vallarai Dysentery, Veterinary
(L.) Urb stomach pain medicine
increases memory
power, ulcer
40 Chamaecrista Leguminosae Gumul Joint pain and
mimosoides (L.) rheumatism
Greene
41 Chromolaena Asteraceae Communist Healing wound
odorata (L.) pacha and burns quickly
R. M. King & cure diarrhoea,
H. Rob. anti-viral,
anti-inflammatory
42 Cinnamomum Lauraceae Karuvapatta Bark used Flavouring
verum J. Presl digestion agent
43 Cissus Vitaceae Parandi Breathing trouble Veterinary
quadrangularis in cattle, stomach medicine
L. ache, dysentery
44 Citrus limon (L.) Rutaceae Elumichai Indigestion, cold
Osbeck and fever
45 Clitoria ternatea Leguminosae Shankupushpam. Antihelminthic
var. ternatea L. and Brain tonic
46 Cocos nucifera Arecaceae Thangai Antiviral and Coir
L. anti-fungal, making
indigestion
47 Colocasia Araceae Chembu Internal
esculenta (L.) haemorrhage
Schott.
48 Cucurbita pepo Cucurbitaceae Sacarakodi Fever, joint pain,
L. dysentery
49 Cuminum Apiaceae Chiragam Anti-helminthic,
cyminum L. stomach pain,
digestive problems
50 Curcuma Zingiberaceae Kasthuri manjal Antiviral,
aromatica Salisb. antifungal, skin
problems
51 Curcuma longa Zingiberaceae Manjal Itching, skin Veterinary
L. problem, antiviral, medicine
anti-fungal,
anti-microbial,
hepatic ulcer
52 Cycas circinalis Cycadaceae Inthpani Stomach pain
L.
(continued)
114 C. V. Jayalekshmi et al.
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
53 Cyclea peltata Menispermaceae Kurupan Stomach pain,
(Lam) Hook. f. child care,
& Thomson anti-inflammatory
54 Cynodon Poaceae Arugampul Epistasis, Cattle feed
dactylon (L.) gastro-intestinal
Pers. problems
55 Cyperus rotundus Cyperaceae Koripul Febrifuge, diuretic
L. and child care
56 Datura metel L. Solanaceae Sipukaichedi Headache,
epilepsy, narcotic
57 Desmodium Leguminosae Orila Asthma, cough,
gangeticum (L.) diuretic, fever
DC.
58 Dioscorea Dioscoreaceae Nurankizhangu. For child care,
pentaphylla L. blood tonic
59 Diplazium Athyriaceae Pannal Used for the
esculentum treatment of
(Retz.) Sw vomiting, asthma
and fever.
60 Eclipta prostrata Asteraceae Kuyuni Laxative, hair
L. tonic,
anti-inflammatory
61 Eleusine Poaceae Ragi Increase strength
coracana (L.) of child bone
Gaertn.
62 Eucalyptus Myrtaceae Eucally Joint pain, cough, Timber
globulus Labill. cure dental
problems,
headache and cold
63 Ficus Moraceae Aalamaram Seminal weakness,
benghalensis L. nervous disorders,
laxative
64 Ficus racemosa Moraceae Atthi Antiseptic, skin
L. diseases
65 Foeniculum Apiaceae Koigerakam Cough, throat pain
vulgare L.
66 Gloriosa superba Colchicaceae Cure bone
L. problems
67 Glycosmis Rutaceae Malakuluki Stomach pain,
pentaphylla dysentery, acidity
(Retz.) DC. and malaria
68 Grewia tiliifolia Malvaceae Lumanmaram Laxative, good for Cattle feed
vahl hair, dysentery,
blood coagulant
(continued)
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 115
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
69 Helicteres isora Malvaceae Keveram Diabetes, ulcer Hair oil
L. and menstrual preparation
pain, stomach
pain, malaria
70 Hemidesmus Apocyanaceae Nannari Blood purifier, Water
indicus (L.) diarrhoea, purification
R. Br. ex Schult. dysentery, stomach
pain increases
memory power
71 Hibiscus rosa - Malvaceae Chemparuthi Hair tonic,
sinensis L. laxative, skin
diseases
72 Ixora coccinea L. Rubiaceae Theei Astringent,
antiseptic
73 Jatropha curcas Euphorbiaceae Kattukotta Anti-inflammatory,
L. wound healing
74 Justicia adhatoda Acanthaceae Adalodakam Bronchitis and
L. acidity, cough,
anti-inflammatory
75 Justicia Acanthaceae Karinochil To treat fractures
gendarussa and dislocated
Burm. f. bones and chronic
indigestion,
dysentery,
rheumatism, fever
and cough.
76 Kyllinga Cyperaceae Muthanga Dysentery and
nemoralis (J. R. white discharge
Forst & Forest).
77 Lantana camara Verbenaceae Paral Abdominal pain, Insecticides
L. rheumatism, tooth
pain
78 Lantana indica Verbenaceae Vettum paral Stomach pain and
Roxb. control vomiting
79 Lawsonia Lythraceae Maruthani Anti-fungal,
inermis L. anti-bacterial,
anti-inflammatory,
antiviral, nail
problems
80 Leucas aspera Lamiaceae Thumbai Headache, Veterinary
(Willd.) Link epilepsy medicine
anti-helminthic,
jaundice,
febrifuge, fever
81 Lycopersicon Solanaceae Takkali Cure pimples on
esculentum Mill. face
(continued)
116 C. V. Jayalekshmi et al.
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
82 Mangifera indica Anacardiaceae Mavu Cure teeth
L. problem,
dysentery
83 Mimosa pudica Leguminosae Thotavadi Kidney stone,
L. diuretic, anti-
inflammatory, eye
problems
84 Momordica Cucurbitaceae Paval Intestinal worms,
charantia L. diabetes
85 Moringa oleifera Moringaceae Murungi Circulatory
Lam. stimulant,
anti-helminthic
86 Murraya koenigii Rutaceae Karivapu Stomach pain,
(L.) Spreng dysentery,
antiprotozoal,
antimicrobial,
antispasmodic,
87 Naravelia Ranunculaceae Kattu kodi Anti-inflammatory,
zeylanica (L.) colic wounds and
DC ulcer
88 Ocimum Lamiaceae Kattutulasi Child care, cough,
americanum L. breathing problem
89 Ocimum Lamiaceae Thiruneetu tulasi Cold, malaria,
basilicum L. antibacterial
90 Ocimum Lamiaceae Tulasi Asthma, cough,
tenuiflorum L. cold
91 Oxalis Oxalidaceae Puliyarali Liver tonic, fever,
corniculata L. hepato-protective
and burning
sensation
92 Passiflora foetida Passifloraceae Pottrichedi Edible
L.
93 Pavetta indica L. Rubiaceae Kollipu Treat poisonous
bite and skin
disease
94 Phyllanthus Euphorbiaceae Kelianelli Jaundice,
amarus dysentery
Schumach. &
Thonn.
95 Phyllanthus Euphorbiaceae Nelli Jaundice,
emblica L. antibiotic
96 Piper betle L. Piperaceae Vettilai Bronchitis,
stimulant, and
antiseptic, cough,
rheumatism
(continued)
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 117
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
97 Piper longum L. Piperaceae Tippili Cough, bronchitis,
asthma
98 Piper nigrum L. Piperaceae Kurmiluku Antiseptic
99 Plectranthus Lamiaceae Iruvali Fever, snake bite,
amboinicus cough, sore throat
(Lour.) Spreng.
100 Plectronia Rubiaceae Kari maram Joint pain,
parvifolia (Roxb) anti-inflammatory,
Benth & Hook. f. fever
exkurz
101 Plumbago Plumbaginaceae Ottachedi Stomach pain,
zeylanica L. anti-helminthic
102 Plumeria alba L. Apocynaceae Vella arali To treat ulcers and
cure nail infections
103 Pongamia Leguminosae Pongum Anti-helminthic,
pinnata (L.) jaundice,
Piere febrifuge,
haemorrhage,
fever
104 Psidium guajava Myrtaceae Koyya Laxative, tonic,
L. dysentery, stomach
pain and gas
trouble
105 Pterocarpus Leguminosae Bark for the Timber
marsupium Roxb. treatment of joint
pain
106 Punica granatum Lythraceae Madalam Good for
L. dysentery,
stomachic and
vomiting
107 Rauvolfia Apocynaceae Povaru Epilepsy,
serpentina (L). gastrointestinal
Benth. exkurz problems, fever
108 Ricinus Euphorbiaceae Kottimuttu Gastro-intestinal Veterinary
communis L. problem, cough, medicine
rheumatism,
stomach pain
109 Rubia cordifolia Rubiaceae Maruchedi Skin disease, ulcer
L. and cough,
anti-inflammatory
110 Santalum album Santalaceae Sandhanam Cooling diuretic,
L. antiseptic, skin
disease, anti-
inflammatory,
antiviral
(continued)
118 C. V. Jayalekshmi et al.
Table 1 (continued)
Sl. Medicinal use/
No. Scientific name Family Common name cures Other uses
111 Sarcostemma Apocynaceae Karumparandi Cure boils
acidum (Roxb)
Voigt.
112 Scoparia dulcis Plantaginaceae Sedative, diuretic,
L. kidney stone
113 Senna Leguminosae Thira Stomach pain,
occidentalis (L.) control ring worms
Link
114 Sida cordifolia L. Malvaceae Maruchedi Fever, cough and
joint pain, nervous
diseases and
rheumatism,
burning and
sensation, cooling,
cough, hair wash
115 Solanum Solanaceae Manathali Wounds, ulcer,
americanum skin disease, fever
Mill.
116 Solanum Solanaceae Siru sundi Jaundice, diuretic
argenteum Dunal
117 Solanum torvum Solanaceae Sundi Respiratory, fever,
Sw. skin ailments,
ulcer, dysentery
118 Solanum Solanaceae Peetachedi Hair wash, cure
verbascifolium L. boils, skin diseases
119 Solanum Solanaceae Nilachundai Stomach pain
xanthocarpum
Schrad. & Wendl
120 Syzygium cumini Myrtaceae Naval Ulcer, diabetes,
(L.) Skeels dysentery
121 Tamarindus Leguminosae Puli Cure jaundice,
indica L. laxative,
astringent, cooling
122 Tephrosia Leguminosae Kolingi Urinary disorder,
purpurea (L.) vomiting
Pers.
123 Trichodesma Boraginaceae Kilukkamtumpa Anti-inflammatory, Veterinary
indicum (L.) rheumatism, medicine
Lehm cough
124 Tridax Asteraceae Sadukkupodukan Anti-inflammatory,
procumbens (L). stomach pain,
L. fever
125 Vitex negundo L. Verbenaceae Notchi Inhaled for cold
and cough
126 Zingiber Zingiberaceae Inji Stomach ache,
officinale Rosc. digestive
problems, gas
trouble
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 119
Out of 126 plant species documented, 123 plants are angiosperms, 2 are pterido-
phytes and 1 is a gymnosperm. Cycas ciricinalis is used as edible plant by tribes in
Parambikulam wildlife sanctuary [14], and it is also used by Malayali tribe of
Servarayan hills for treating leucorrhoea [15]. Angiopteris evecta and Diplazium
esculentum were the Pteridophytes documented in this study. Medicinal importance
of both these plants were already documented [16, 17].
Out of 126 plant species, 37% of plants were herbs, 23% shrubs, 21% were trees
and 19% climbers. Figure 2 depicts the habit of plants. Most of the plants were used
to cure digestive system disorders (56), followed by fever (38) and skin disorders
(22). Figure 3 depicts the number of plants used for each disease category.
19%
37%
21%
23%
No of plants used
Kidney stone
Rheumatism and joint pain
Healing of wounds
Malaria
Jaundice
Anti-inflammatory activity
Hair care
Fever
Urinary disorders
Epilepsy
Sexual and menstrual disorders
Skin diseases
Snake bite
Respiratory disorders
Digestive system Disorders
0 10 20 30 40 50 60
Eight plants were used to treat jaundice, three plants to treat malaria, two plants
to treat kidney stones and three plants to treat epilepsy. The most commonly cited
plants were Hibiscus rosa-sinensis, Leucas aspera and Ricinus communis by 4
informants. Hibiscus possessed analgesic, antipyretic, anti- asthmatic, anti-
inflammatory, antioxidant, antifungal, antimicrobial and hair growth promoting
activity [18]. In the present study, Irula tribes mainly use Hibiscus rosa-sinensis for
hair care and also as laxative. Antioxidant, antibacterial, antifungal and cytotoxic
activities of Leucas aspera were already reported [19, 20]. Ricinus communis pos-
sessed antidiabetic, antimicrobial, antifungal, analgesic, insecticidal, antitumour,
antiasthmatic, antinociceptive, antifertility and bone degeneration activity [21].
Informant consensus factor or Fic value is high for kidney stone (0.6) followed
by fever (0.412). For kidney stone, 2 plants were cited with four used reports. High
Fic value indicates that a few taxa are used by the community for the treatment of a
particular disease category [22]. The Fic values for different disease categories are
given in Table 2.
4 Conclusion
The present study documented 126 plants used by the Irula tribal settlement. 29
informants participated in the survey, among them some were traditional healers.
Irula tribes use a wide variety of plants to treat various diseases. Documentation of
this traditional knowledge is essential as the tribal people do not share their knowl-
edge with a third person. Further studies on these plants may lead to the production
of new drug molecules.
Ethnomedicinal Plants Used by Irula Tribal Settlement of Attappady in Palakkad… 121
Acknowledgements The authors are thankful to the Principal, teachers, staff and students of
Department of Botany, Government Victoria College, Palakkad. The authors are thankful to the
informants of Irula tribal settlement Attappady for sharing their valuable traditional knowledge.
The authors are thankful to UGC and CSIR for financial assistance.
References
1. Rajith N, Ambily D, Dan VM, Devi PS, George V, Pushpangadan P (2012) A survey on eth-
nomedicinal plants used for menstrual disorders in Kerala. Indian J Tradit Knowl 11:453–460
2. Rehecho S, Uriarte-Pueyo I, Calvo J, Vivas LA, Calvo MI (2011) Ethnopharmacological sur-
vey of medicinal plants in Nor-Yauyos, a part of the Landscape Reserve Nor-Yauyos-Cochas,
Peru. J Ethnopharmacol 133:75–85
3. Tahira B, Mushtaq A, Rsool BT, Niaz MT, Rukhsana J, Saeed-Ur R, Shazia S, Muhammad
Z, Ghulam Y (2014) Ethnobotany of medicinal plants in district Mastung of Balochistan
province-Pakistan. J Ethnopharmacol 157:79–89
4. Rahaman CH, Karmakar S (2014) Ethnomedicine of Santal tribe living around Susunia hill of
Bankura district, West Bengal, India: the quantitative approach. J Appl Pharm Sci 5:127–136
5. Phillips O, Gentry AH, Reynel C, Wilkin P, Galvez-Durand BC (1994) Quantitative ethno-
botany and Amazonian conservation. Conserv Biol 8:225–248
6. Chathukulam J, Reddy MG, Rao PT (2012) An assessment and analysis of tribal sub-plan
(TSP) in Kerala. Research Unit for Livelihoods and Natural Resources
7. Sulochana A, Raveendran D, Krishnamma A, Oommen O (2015) Ethnomedicinal plants used
for snake envenomation by folk traditional practitioners from Kallar forest region of South
Western Ghats, Kerala, India. J Intercult Ethnopharmacol 2015(4):47–51
8. Haseena VA (2015) Poverty and livelihood problems among the scheduled tribes in Kerala-a
study on Attappady. J Poverty Invest Dev 14:94–101
9. Varghese A, Nagaraj P (2012) A study on the tribal culture and folklore of Attappady. Galaxy
Int Multidiscip Res J 1:1–8
10. Alex A, Vidyasagaran K, Prema A, Kumar AVS (2016) Analyzing the livelihood opportunities
among the tribes of the Western Ghats in Kerala. Stud Tribes Tribals 14:11–17
11. Veena George M, Christopher G (2017) Nutritional value of selected wild edible leaves used
by tribal communities of Attappady, Southern Western Ghats. Int J Food Sci Nutr 2:126–132
12. Hoeschele W (2000) Geographic information engineering and social ground truth in Attappadi,
Kerala State, India. Ann Assoc Am Geogr 2000(90):293–321
13. Heinrich M, Ankli A, Frei B, Weimann C, Sticher O (1988) Medicinal plants in Mexico: heal-
ers’ consensus and cultural importance. Soc Sci Med 47:1859–1871
14. Yesodharan K, Sujana KA (2007) Wild edible plants traditionally used by the tribes in the
Parambikulam Wildlife Sanctuary, Kerala, India. Indian J Nat Prod Resour 6:74–80
15. Udayan PS, George S, Tushar KV, Balachandran I (2006) Medicinal plants used by the Malayali
tribe of Servarayan Hills Yercad Salem District Tamil Nadu India. Zoos’ Print J 21:2223–2224
16. Semwal P, Painuli S, Painuli KM, Antika G, Tumer TB, Thapliyal A, Setzer WN, Martorell
M, Alshehri MM, Taheri Y (2021) Diplazium esculentum (Retz.) Sw.: Ethnomedicinal, phyto-
chemical, and pharmacological overview of the Himalayan Ferns. Oxidative Med Cell Longev
2021:1–15
17. Udayan PS, George S, Tushar KV, Balachandran I (2005) Medicinal plants used by the Kaadar
tribes of Sholayar forest Thrissur district, Kerala. Indian J Tradit Knowl 4:159–163
18. Missoum A (2018) An update review on Hibiscus rosa sinensis phytochemistry and medicinal
uses. J Ayurvedic Herb Med 4:135–146
122 C. V. Jayalekshmi et al.
19. Babu A, Mohamed MSN, Jaikumar K, Anand D, Saravanan P (2016) In-vitro antifungal activ-
ity of leaf extracts of Leucas aspera and Leucas zeylanica. Int J Pharm Sci Res 7:752–756
20. Prajapati MS, Patel JB, Modi K, Shah MB (2010) Leucas aspera: a review. Pharmacogn
Rev 4:85–87
21. Jena J, Gupta AK (2012) Ricinus communis Linn: a phytopharmacological review. Int J Pharm
Pharm Sci 4:25–29
22. Heinrich M (2000) Ethnobotany and its role in drug development. Phytother Res 14:479–488
Folk Medicine of Chittur Taluk
in Palakkad District, Kerala, India
1 Introduction
Plants are widely used as a source of food, fodder, fuel, medicine, etc. India is rich in plant
diversity. The people of India mainly depend on plants to treat many diseases. Our Vedas,
Rigveda and Atharvaveda and ancient books like Charakasamhita, Sushruthasamhitha,
Dhanwanthari nighantu are the important sources of information on medicinal plants [1].
Traditional medicinal practices are still widely used in various parts of India.
According to WHO, about 80% people in developing countries depend on
medicinal plants for their primary health care. In India, Ayurveda, Yunani, Siddha,
folk medicine and tribal medicine depend mainly on plants to treat various diseases.
Kerala is the southernmost state of India with a vast diversity of medicinal plants.
The people of Kerala used plants as a source of medicine from very ancient times.
Folk medicine commonly called ‘Nattu chikitsa’ originated around the thirteenth
century A.D [2]. Folk medicine used around 25,000 plant-based formulations [3].
Folk medicine is still widely practiced in Kerala as it is safe and cheap. Palakkad
district is located in the eastern part of Kerala and has no sea coast. Chittur taluk is
The original version of this chapter was revised. The correction to this chapter is available at
https://doi.org/10.1007/978-3-031-28780-0_65
C. V. Jayalekshmi
Research Scholar, Post Graduate and Research Department of Botany, Government Victoria
College Palakkad, Palakkad, Kerala, India
S. Reshma
Post Graduate and Research Department of Botany, Government Victoria College Palakkad,
Palakkad, Kerala, India
V. Suresh (*)
Assistant Professor, Post Graduate and Research Department of Botany, Government Victoria
College Palakkad, Palakkad, Kerala, India
The present study was conducted in Chittur taluk of Palakkad district. Frequent field
visits were conducted to the study area for collection of information on medicinal
plants used in folk medicine.
2.1 Study Area
2.2 Collection of Data
Survey was used to collect information regarding the use of plants in folk medicine.
Fifty-six informants from 15 villages were interviewed. The data were recorded in
survey sheets. Frequent field visits from February to July 2020 were conducted in
study areas for data collection. People within the age range of 30–80 years were
interviewed. Pothundy, Kuriyallur, Pottakkalpadam, Mettupalayam, Chunkam,
Kallandichalla, Thekkechalla, Ayyappankavu, Panniperunthala, Adichera,
Kattutheruvu, Athikode, Annikode, Arampadam and Attancheri are the villages
selected for the present study. Common name of plants, plant part used, mode of
delivery and curative properties of these plants were documented.
The plants documented in this study were collected and identified using available
taxonomic literatures. The voucher specimens are deposited in Government Victoria
College Herbarium.
2.3 Analysis of Data
FicValue N ur N t / N ur 1
Folk Medicine of Chittur Taluk in Palakkad District, Kerala, India 125
The present study deals with documentation of medicinal plants used in folk medicine
of Chittur taluk, Palakkad district, Kerala. The study reported 212 plants. The most
commonly cited families were Leguminosae, Euphorbiaceae, Apocynaceae by and
Malvaceae. Out of 212 plants, 75 plants were herbs, 52 plants were shrubs, 52 plants
were trees and 33 plants were climbers. Figure 2 depicts the habit of plants cited in the
study. The plants cited in this study are represented in Table 1 with their botanical
name, family, local name, medicinal value and mode of administration. Most fre-
quently used plant part is leaves followed by roots, flowers, fruits, rhizomes, seeds and
whole plant. Photographs of some plants cited in the study are given in Plate 1.
The most common ailments were fever, stomach problems and skin problems.
Seventy-three plants are used to treat digestive system disorders, 8 plants are used
to treat respiratory system disorders, 21 plants are used to treat snake bite, insect
bite and dog bite, 52 plants are used to treat skin diseases, 44 plants possess wound
126 C. V. Jayalekshmi et al.
Habit of plants
16%
36%
24%
24%
healing properties and 65 plants are used for general disorders, etc. Figure 3 depicts
the number of plants used for various diseases.
Single plant is used for more than one disease. For example, Tinospora sinensis
used for fever, kidney problem and diabetes, Sida cordifolia used to treat diarrhoea,
fever, urinary infection and skin disease, Ricinus communis was used to treat skin
diseases, joint pain, ear pain and constipation and Achyranthes aspera used for
fever, cough, cold and scorpion bite. The different parts of the same plant have dif-
ferent properties. For example, in the case of Ocimum tenuiflorum, the leaves are
used to treat throat infection, root extract was used to treat cough and the seeds were
used to treat eye irritations. Similarly, Sida cordifolia root extract is used to treat
fever and leaf extract was used for skin warts and wounds.
Tinospora sinensis and Azadirachta indica were the commonly cited plants in
this study followed by Curcuma longa. Anti-inflammatory, anti-oxidant, immuno-
modulatory, hepatoprotective and anti-leishmanial properties of T. sinensis were
already reported [6–10]. Anti-inflammatory, antipyretic, hepatoprotective, anti-
microbial, anti-cancer, immunomodulatory, anti-candidal properties of A. indica
were also reported [11–13]. These properties indicate the medicinal importance of
these plants.
Five plants were used to treat jaundice and three plants were used to treat malaria.
All the five plants cited in this study were used for treating jaundice are already
reported. Ethnobotanical studies from Maharashtra reported that Luffa acutangula
fruits are used to treat jaundice [14]. Ethnobotanical studies from Bangladesh
reported the use of Asparagus racemosus to treat jaundice [15]. Studies on hepato-
protective plants used by the tribes of Wayanad, Malappuram and Palakkad reported
the use of Centella asiatica to treat jaundice [16]. The tribes of Akole taluk of
Maharashtra use Sesbania grandiflora to treat jaundice [17]. Studies on folk medi-
cine from Chittur district, Andhra Pradesh, reported the use of Phyllanthus amarus
Table 1 Plants cited in the study
Sl. Common name of
No plant Scientific name Family Used to cure Mode of delivery
1 Vendakka Abelmoschus esculentus (L.) Malvaceae (a) To improve memory power (a) Fruits used as a vegetable in daily
Moench and for abdominal disorders food
2 Kunni Abrus precatorius L. Leguminosae (a) Best to treat swellings and (a) Leaves mixed with coconut oil then
joint pain placed
(b) Treats inflammations (b) Leaf and stem extract are used
3 Cheeppika Abutilon indicum (L.) Sweet Malvaceae (a) Used as anti-inflammatory (a) Dried powder
agent
4 Munja Acalypha fruticosa Forssk. Euphorbiaceae (a) Protect new born babies (a) Leaves and flowers boiled in water
from cold and cough then to bath babies in the cooled water
5 Kuppameni Acalypha indica L. Euphorbiaceae (a) For the treatment of skin (a) Take juice extract from leaves and
allergies mixed with lime then applied on the skin
(b) Treat rheumatism (b) Leaf juice with coconut oil is applied
(c) Cure wounds (c) Powdered leaves can be applied
6 Sapota Achras sapota L. Sapotaceae (a) Fruits provide good diet (a) Ripened fruits are eaten
7 Kadaladi Achyranthes aspera L. Amaranthaceae (a) Cures cough and throat (a) Ash of spike is mixed with honey and
infection in infants and also applied
cure toothache (b) Paste made from spike and applied
(b) Against scorpion bite
Folk Medicine of Chittur Taluk in Palakkad District, Kerala, India
8 Venapacha Acmella radicans (Jacq) Asteraceae (a) Treat toothache, gum (a) Plant extract used as mouthwash
R. K. Jansen. infections and also for gum
swellings
10 Cherula Aerva lanata (L.) Juss. Amaranthaceae (a) The plant used for the (a) Decoction prepared with the root of
treatment of diarrhoea in cyperus and fennel seed
children (b) The extract of whole plant is mixed
(b) Cures bleeding during with milk
pregnancy time (c) Decoction of plant taken
(c) Treat urinary infection
127
(continued)
Table 1 (continued)
128
is applied
41 Pachamulak Capsicum annum L. Solanaceae (a) Used as an appetizer (a) Fruit as an ingredient of food
42 Uzhinja Cardiospermum halicacabum Sapindaceae (a) Treatment of ulcer (a) Leaf extract is applied on wounds
L. To cure fever (b) Leaves boiled in water and drink the
(b) Improves hair growth water
(c) Plant extract mixed with hibiscus
leaves and applied
43 Papaya Carica papaya L. Caricaceae (a) Remove intestinal worms (a) Ripe or unripe fruits take on empty
and treat kidney stone stomach
(continued)
131
Table 1 (continued)
132
72 Poovamkurunthila Cyanthillium cinereum (L.) Asteraceae (a) Treatment of red eye and (a) Leaf extract mixed in honey then
H. Rob. irritations applied as eye drops
73 Karukapullu Cynodon dactylon (L.) Pers. Poaceae (a) To improve blood volume (a) Daily intake of plant extract
(b) Treatment of bleeding (b) Plant extract applied on nose
from nose (c) Plant extract mixed with cow milk
(c) Treatment of seasonal cleft then applied on lip
lip
(continued)
135
Table 1 (continued)
136
118 Muthanga Kyllinga nemoralis Cyperaceae (a) Relief from acidity (a) Prepared decoction with the plant and
(J.R.Forst. & G.Forst.) problems taken
Dandy ex Hutch. & Dalzel (b) To treat cold and fever (b) Whole-plant extract is used
(c) Treat dysentery (c) Fresh tubers made into paste and taken
with rice
119 Avara Lablab pupureus (L) Sweet. Leguminosae (a) To relieve internal fever (a) Decoction of leaves and seeds
(b) Treatment of vomiting consumed
(b) Paste of seeds consumed
120 Poochedipu Lantana camara L. Verbenaceae (a) Quick healing of wounds (a) Apply the extract from leaves on it
(continued)
141
Table 1 (continued)
142
135 Thottavadi Mimosa pudica L. Leguminosae (a) Relief to chest pain (a) Root paste consumed
(b) To control diabetes (b) Juice from whole plant is consumed
(c) Control excessive bleeding (c) Plant juice takes along with honey
during menstruation (d) Leaf juice mixed with coconut water
(d) Cure breathing troubles in and taken
children
136 Nalumanipu Mirabilis jalapa L. Nyctaginaceae (a) Treat wounds and (a) Leaf juice is applied on the wounds
inflammations
(continued)
143
Table 1 (continued)
144
150 Poochapalam Passiflora foetida L. Passifloraceae (a) Treating digestive (a) Ripe fruits are taken
problems including diarrhoea
151 Vellathechi Pavetta indica L. Rubiaceae (a) Relief from wound pain (a) Crushed leaves applied on wounds
152 Thathamachedi Pedilanthus tithymaloides Euphorbiaceae (a) Treat insect sting (a) Plant latex is applied externally
(L.) Poit.
153 Mashithandu Peperomia pellucida (L.) Piperaceae (a) Treatment of fire burns (a) Plant juice applied on the affected skin
kunth
(continued)
145
Table 1 (continued)
146
Pierre. (b) Quick healing of wounds (b) Paste made with leaves and seeds is
and relieve pain applied
170 Karincheera Portulaca oleracea L. Portulacaceae (a) Improve vision power (a) Leaves cooked and taken
(continued)
147
Table 1 (continued)
148
vegetable
184 Kurunthotti Sida acuta Burm. f. Malvaceae (a) Relieve from fever and (a) Whole-plant extract is prepared into a
treats asthma decoction
(continued)
149
Table 1 (continued)
150
(b) For good health of (b) Daily intake of seeds with milk
pregnant women (c) Oil prepared with seeds mixed in
(c) To maintain black and coconut oil then heated and applied
thick hair
198 Poovarasu Thespesia populnea (L.) Sol. Malvaceae (a) Treatment of psoriasis (a) Oil prepared by heating bark in
ex Correa coconut oil then applied on skin
(continued)
151
Table 1 (continued)
152
4 Conclusion
The present study aimed to document medicinal plants used in the folk medicine of
Chittur taluk, Palakkad district, Kerala. Traditional remedies used for the treatment
of various diseases are considered to be important in the primary health care of
people. Most of the people in village areas still depend on plants for first aid reme-
dies and to treat common health problems such as fever, stomach ache and some
simple ailments. Majority of the informants were aged above 50 years indicating
that younger generations are unaware of many medicinal plants. From the survey,
212 plants with different medicinal properties were recorded. The valuable
156 C. V. Jayalekshmi et al.
knowledge about medicinal plants was unwritten and depleted significantly from
generation to generation. So this documentation will help to save the traditional
medicines of Chittur taluk.
Acknowledgements The authors are thankful to the principal, teachers, staff and students of
Department of Botany, Government Victoria College, Palakkad. The authors are thankful to the
informants of Chittur taluk, Palakkad for sharing their valuable traditional knowledge. The first
author is thankful to UGC for financial assistance.
References
1. Rajith NP, Navas M, Thaha AM, Manju MJ, Anish N, Rajasekharan S, George V (2010) A
study on traditional mother care plants of rural communities of South Kerala. Indian J Tradit
Knowl 9:203–208
2. Rasiya BA, Nayar TS (2011) Plants used for natal healthcare in folk medicine of Kerala, India.
Indian J Tradit Knowl 10:523–527
3. Verma S, Singh SP (2008) Current and future status of herbal medicines. Vet World 1:347–350
4. Heinrich M (2000) Ethnobotany and its role in drug development. Phytother Res 14:479–488
5. Bulut G, Tuzlaci E (2013) An ethnobotanical study of medicinal plants in Turgutlu (Manisa-
Turkey). J Ethnopharmacol 149:633–647
6. Lam SH, Chen PH, Hung HY, Hwang TL, Chiang CC, Thang TD, Kuo PC, Wu TS
(2018) Chemical constituents from the stems of Tinospora sinensis and their bioactivity.
Molecules 23:2541
7. Manjrekar PN, Jolly CI, Narayanan S (2000) Comparative studies of the immunomodulatory
activity of Tinospora cordifolia and Tinospora sinensis. Fitoterapia 71:254–257
8. Nagarkar B, Kulkarni R, Bhondave P, Kasote D, Kulkarni O, Harsulkar A, Jagtap S (2013)
Comparative hepatoprotective potential of Tinospora cordifolia, Tinospora sinensis and Neem-
guduchi. Br J Pharm Res 3:906
9. Narkhede AN, Jagtap SD, Kasote DM, Kulkarni OP, Harsulkar AM (2014) Comparative
immunomodulation potential of Tinospora cordifolia (Willd.) Miers ex Hook. F., Tinospora
sinensis (Lour.) Merrill and Tinospora cordifolia growing on Azadirachta indica A. Juss.
Indian J Exp Biol 52:808–813
10. Singh N, Kumar A, Gupta P, Chand K, Samant M, Maurya R, Dube A (2008) Evaluation of
antileishmanial potential of Tinospora sinensis against experimental visceral leishmaniasis.
Parasitol Res 102:561–565
11. Lloyd AC, Menon T, Umamaheshwari K (2005) Anticandidal activity of Azadirachta indica.
Indian J Pharm 37:386–389
12. Malar TJ, Antonyswamy J, Vijayaraghavan P, Kim YO, Al-Ghamdi AA, Elshikh MS, Hatamleh
AA, Al-Dosary MA, Na SW, Kim HJ (2020) In-vitro phytochemical and pharmacological bio-
efficacy studies on Azadirachta indica A. Juss and Melia azedarach Linn for anticancer activ-
ity. Saudi J Biol Sci 27:682–688
13. Okpanyi SN, Ezeukwu GC (1981) Anti-inflammatory and antipyretic activities of Azadirachta
indica. Planta Med 41:34–39
14. Mulay JR, Sharma PP (2003) Plants used in treatment of jaundice by folklore of Ahmednagar
district, Maharashtra, India. Sci Res Rep 3:216–222
15. Rahim ZB, Rahman MM, Saha D, Hosen SZ, Paul S, Kader S (2012) Ethnomedicinal plants
used against jaundice in Bangladesh and its economical prospects. Bull Pharma Res 2:91–105
16. Asha VV, Pushpangadan P (2002) Hepatoprotective plants used by the tribals of Wynadu,
Malappuram and Palghat districts of Kerala, India. Anc Sci Life 22:1–8
Folk Medicine of Chittur Taluk in Palakkad District, Kerala, India 157
17. Wabale AS, Petkar AS (2005) Ethnomedicinal plants used against jaundice by the tribals of
Akole taluka (MS). J Phytol Res 18:259–261
18. Reddy KR (1988) Folk medicine from Chittoor District, Andhra Pradesh, India, used in the
treatment of jaundice. Int J Crude Drug Res 26:137–140
19. Ankrah NA, Nyarko AK, Addo PG, Ofosuhene M, Dzokoto C, Marley E, Addae MM, Ekuban
FA (2003) Evaluation of efficacy and safety of a herbal medicine used for the treatment of
malaria. Phytother Res 17:697–701
20. Namdeorao Tankar A. Pharmacognostic phytochemical pharmacological evaluation of differ-
ent parts of callicarpa tomentosa ll and formulation development of bioactive extract. 2019
21. Tan LTH, Lee LH, Yin WF, Chan CK, Abdul KH, Chan KG, Goh BH (2015) Traditional uses,
phytochemistry, and bioactivities of Cananga odorata (Ylang-Ylang). Evid Based Complement
Alternat Med 2015:1–30
Checklist Flora of Sunderdhunga Valley,
Western Himalaya, with Emphasis
on Ethno-Medicinal Plants
1 Introduction
It is worth to explore any area with wide range of forest types and to identify eco-
nomically and medicinally important plants found there. Further, the reserve forests
and territorial forests are preserved for specific purposes, such as proper mainte-
nance of the wildlife in that area and prevention of human interference through
activities, like deforestation, cultivation and grazing of animals. It is a rewarding
experience to make available a systematic account of the floristic wealth of such
region or area. Moreover, the knowledge about the plants in any region is essential
with the increasing consciousness of people about the environment and its impact
on living organisms in general. Hence, it is very important to conserve our plant
wealth or diversity for the major role in providing food, fuel, fibres, medicine, shel-
ter, maintaining a stable environment and to study the scientific value of plants. This
is useful in assessing the periodic changes in flora that may occur in the study area.
With this conviction the Flora of Sunderdhunga and adjoining is being evaluated.
The Botanical Survey of India under the Ministry of Environment and Forest and
Climate Change has been allotted a project to document the floristic account of vari-
ous groups of wild plants of our country. The Sunderdhunga and adjoining in
Uttarakhand is one such area located in the Western Himalayas and together with
Eastern Himalaya which is one of the largest centres of endemism in India is among
the 34 biodiversity hotspots of the world [1]. According to Myers et al. [2] biodiver-
sity ‘hotspots’ are the regions containing a high proportion of global biodiversity in
a small area. Also biodiversity hotspots can be defined as areas having exceptional
concentration of endemic taxa. The Sunderdhunga and adjoining is situated in the
Bageshwar district of Uttarakhand a newly formed state from Uttar Pradesh.
The Sunderdhunga glacier and its surrounding areas encompass a rich biodiversity
of both flora and fauna and it is located at Bageshwar district in the Western
Himalaya. It covers of rich Sub-tropical Quercus forest, Himalayan moist temperate
forest, Himalayan dry-temperate forest, Sub-alpine forest and moist alpine forest.
The area is very rich in medicinal plants many of which form base for certain life-
saving drugs. It is a part of Nanda Devi Biosphere Reserve, which is one of the
World Heritage Sites, famous for its pristine and exceptional beautiful landscape.
The landscape of the study area is rolling large meadows, criss-cross dancing rivers
and streams, high-altitude lakes and snow clad peaks. The elevation varies from
1300 to 6050 m. It has unique social and cultural style of the area altogether repre-
sents sole scenic beauty which is unparalleled in the entire Himalayas. Also, it has
a great tourism potential especially glacier regions of Maiktoli glacier, Sukhram
glacier, etc. The area houses a lot of endangered fauna, e.g. Snow leopard, Brown
bear, Black bear, Bharal, Musk deer, Monal pheasant, etc. It is a significant compo-
nent of one of the snow leopard conservation areas in India. In addition, the India
Eco-development Project has also been implemented in the villages surrounding the
area in order to reduce the human pressures on wildlife habitat and also various
development programmes, such as research, monitoring, training and conservation,
are taken up by the Forest Department as well as other Government agencies to
meet various issues.
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 161
The present study was undertaken to explore this under-explored area which is
endowed with a rich floristic diversity. Extensive and intensive plant explorations
were undertaken to assess and prepare the floristic account and plant wealth of
Sunderdhunga and adjoining.
162 R. Manikandan et al.
According to the report of FAO (1993) shows the rate of deforestation is esti-
mated to be 15 million hectares per annum. Besides, the Uruguay Round of the
General Agreement on Trade and Tariffs (GATT) has led to greater vulnerability of
forest elements owing to market prices, new seed varieties and patenting laws.
There is an alarming trend of forests transforming into agro industries. In addition,
introduction of exotic species of flora whether accidentally or intentionally in the
forests threatens local biodiversity. Recently, Pitman & Jorgensen [3] found that the
number of species unique to each country is a rough guide to the number that is
threatened and nearly half of the world’s plants could be close to extinction.
According to National Remote Sensing Agencies (NRSA), the natural forests
support the maximum biodiversity, which are depleting at a very fast rate. Our coun-
try too is losing its natural forests at the rate of 1.3 m ha every year [4]. Ahmedullah
& Nayar [5] have estimated about 1500 species accounting for about 10% of the
flowering plants of India are threatened. Thus, to protect and conserve the remain-
ing 90% of the flowering plants, a judicious management and innovative methods of
conservation are required. India is facing an alarming rate of loss of floristic diver-
sity particularly due to declining forest cover and species.
Major problems faced in the present study area are cattle grazing, forest fire, dif-
ficulty in managing wildlife corridors and human interference mostly due to tour-
ism. As the reserve forests/territorial forests are the prime centres of conservation
and diversity, scientific information on all taxa needs to be collected and docu-
mented. Therefore, it is very important to make an assessment of the existing floris-
tic diversity. Considering these reasons, Sunderdhunga valley and adjoining was
selected for an assessment of floristic diversity.
1.5.1 Past Work
Hooker & Thomson [6] in their ‘Flora Indica’ reported quite comprehensive infor-
mation on the flora of Garhwal Himalaya which includes the present study area.
Hooker and his collaborators have dealt with many species in The Flora of British
164 R. Manikandan et al.
India [7]. The work of Atkinson [8] on Flora of the Himalayas with special refer-
ence to Kumaon, Garhwal, Nepal and Tibet and Duthie’s [9] Flora of Garhwal
Himalaya can be conveniently considered as one of the eminent contributions in
the field.
Some scholars like Deva & Naithani [10], Garg [11], Aswal et al. [12], Dangwal
[13] and Nautiyal [14] have made efforts to study the flowering plants of the Garhwal
Himalaya in terms of certain families or groups of a given geographical entity.
Important information can be derived from Flowers of the Himalaya [15],
Blossoming Garhwal Himalaya [16], Flowers of Himalaya [17], Flowers of West
Himalaya [18] and Naithani’s Flora of Chamoli [19]. Other important unpublished
works include Manis Kandwal’s [20] Grass Flora of Uttarakhand. Many other bot-
anists, like Duthie [21, 22]; Issar & Uniyal [23]; Khullar [24], Osmaston [25]; Pande
[26–29]; Pangey, et al [30]; Rai et al. [31]; Randhwa [32]; Rau [33, 34]; Rawat [35];
Rawat & Sharma [36]; Singh et al. [37] and Uniyal et al. [38], have contributed for
the Flora of Kumaon Himalaya.
1.5.2 Present Work
Intensive and extensive surveys have been undertaken periodically along different
topographic and climatic gradients within the potential area of Sunderdhunga and
adjoining, in order to achieve the above said objectives. The duration of each study
tour ranged from 15 to 20 days. While carrying out the assessment of floristic diver-
sity, efforts will be made to collect the plants in all seasons. The field observations
such as habit, habitat, flower colour, odour, distribution, along with local names and
uses, if any gathered from the tribes of the area has to be noted in the field itself and
any details which cannot be deduced from a herbarium specimen.
2.1 Herbarium Study
After returning from the field, plant specimens, which have been tentatively identi-
fied in the field, have been carefully and critically studied, before these could be
processed through conventional methods of drying, poisoning, mounting, stitching
and labelling [39, 40].
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 165
2.2.1 Geology
The entire landform lies in the Himalaya stratigraphically zone and is in the vicinity
of main central propelled, which shows the serious impact of tectonic movements.
As a result, rocks are not only tightly folded but also broken into sheets and arranged
one over the other, forming tall heaps. The internal configurations of these sheets
are complicated cleavage and have developed inside thrust fold valley, which seri-
ously affected during heavy rains and earth quake. The whole area is prone to large-
scale landslide. The elevation of the valley varies from 1300 to 6050 m.
2.2.2 Rocks
This area falls under Sub-Himalayan range; however, some parts of upper area
extend till Greater and Mid-Himalayan regions. The forest sections of this forest
regions extend between the longitudes of 79°28′30″ E to 80°29′30″ E and the lati-
tudes of 29°58′45″ N to 30°9′15″ N. This region consists of three main strips, which
are west, north-west, east and south-east, respectively. The whole terrain is broken
by numerous streams and nallahs. All aspects of gradients are occurs in the present
study area. The greater part being steep to precipitous and only a very few gentle
slopes occur in this area. Further, most of the valley or study area is covered by
metamorphic rocks. These rocks principally comprise quartzite, lime stone, magne-
tite, base metal and slate.
2.2.3 Soil
The structure and composition of the soil have a marked influence on the nature of
vegetation along with other factors, like climate, rainfall and biotic. The soil varies
from sandy loam to stiff clay depending on the nature of the schist.
166 R. Manikandan et al.
2.2.4 Climate
The valley experiences greatly varying climate conditions, mainly due to large vari-
ation in altitudes, which is natural to encounter diversified climates in this forest
region. Severe cold can be felt during winters and regions higher than 1520 m
receive snowfall too. At times the lower parts also go through snowfall. The dura-
tion of December to February is of extreme cold in this region and mornings are full
of dense fog.
2.2.5 Rainfall
The valley receives rainfall from both monsoon and winter seasons. The monsoon
starts from the middle of June to the second week of September. The heaviest rain-
fall occurs during July and August.
2.2.6 Temperature
Three seasons, like summer, monsoon and winter, can be recognized in the present
study area. The monsoon starts from the middle of June and last till second week of
September. The peak rainfall occurs during July and August. The entire area of the
valley is subjected to light to heavy snowfall for about 3–4 months from December
to March in winter seasons. The summer season starts from March to the second
week of June. The temperature during summer goes up to 35 °C in lower parts of
Loharkhet and Song valley.
2.2.7 Water Sources
The Sunderdhunga and Pindari are the main rivers in the valley and they have sev-
eral tributaries and also a large number of perennial streams and nallahs, most of
which are snow fed.
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 167
2.2.8 Winds
The winds are light moderate, but sometimes, high and exposed ridges do experi-
ence strong winds.
2.3 Ethnic Groups
According to Lynch [43] in India, out of the human population of over 1 billion,
64% of the rural population and 100 million tribes depend on the forests for their
sustenance. Over 90 million cattle graze inside the forests [44]. According to
Regional Wood Energy Development Programme in Asia, the firewood consump-
tion in India is 1,73,412 k. tons with 62% derived from the forests [45]. According
to a survey carried out in the mid-1980s, over 65% of protected areas were charac-
terized by human settlement and resource use [46]. In the past two decades, these
factors have drawn the attention of policy makers who shifted their main concern
from protecting timber supplies to issues concerning local livelihoods, involvement
of local people in the forest management while formulating conservation agenda.
The inhabitants of Bhotia of Bageshwar district are geographically in Kumaon
region, but they are quite different from Garhwalis in their social and cultural group.
The distinct and unique feature of hill culture, polyandry, was in vogue in Bhotia
areas adjoining. Their houses are made up of Deodar wood and stone, which is three
to four storeyed. In these multi-storeyed houses, the lower one is used for cattle and
fodder storage and the uppermost one was used by human habitation. These storeys
are internally linked for adopting snowbound winters, when it is not possible to go
out of the house and away from hearth. Most of them engaged in marginal agricul-
tural and seasonal employment in various departmental works are the only source of
livelihood.
2.4 Vegetation
(a) Flora
The vegetation of the study area has been classified mainly as Sub-tropical
Quercus forest, Himalayan moist temperate forest, Himalayan dry-temperate forest,
Sub-alpine forest and moist alpine forest [47]. Further, the structural diversity and
representativeness of forest vegetation of Kumaun Himalayas studied by many
workers like Dhar et al. [48], Rau [49], Rawal et al. [50], Rikhari et al. [51], Singh
& Singh [52–54], Tewañ & Singh [55] and Upreti et al. [56].
168 R. Manikandan et al.
2.4.1 Floristic Diversity
During the above said period the following species were collected and identified
from the study area itself:
Acer caesium, Aesculus indica, Alnus nepalensis, Androsace sp., Anemone spp.,
Orchis nana, Arisaema spp., Arnaria ciliolata, Arnebia benthamii, Astragalus pin-
dreensis, Betula utilis, Brassica juncea, Calanthe tricarinata, Caltha palustris,
Cannabis sativa, Corydalis cashmeriana, C. stracheyi, Cupressus torulosa,
Fragaria nubicola, Gagea elegans, Gentiana spp., Indigofera spp., Iris kumaonen-
sis, Juglans regia, Lloydia serotina, Micromeria biflora, Ophiopogon intermedius,
Poa sp., Polygonum spp., Potentilla atrosanguinea, Primula denticulata, P. edge-
worthii, Primula spp., Rhododendron arboreum, R. campanulatum, R. lepidotum,
Rosa spp., Sinopodophyllum hexandrum, Schisandra grandiflora, Taxus wallichi-
ana, Trifolium spp., Vincetoxicum hirundinaria, Viola spp., etc.
1. Sub-tropical Quercus forest
This type of forests occurs in Song and Loharkhet areas where altitude ranges
up to 1300 m.
The upper storey comprises Acer oblongum, Alnus nitida, Lyonia ovalifolia,
Pyrus pashia, Quercus leucotrichophora, Rhododendron arboreum, etc., and the
lower storey comprises Chrysopogon fulvus, Debregeasia salicifolia, Flemingia
macrophylla, Heteropogon contortus, Prinsepia utilis, Rhamnus virgatus, Rubus
ellipticus, Woodfordia fruticosa, etc.
2. Himalayan moist temperate forest
(a) Ban oak forests (Quercus leucotrichophora forests):
This type of forests occurs in Dhakuri range where altitude ranges up to
2250 m. The forests are generally well-stocked and main associates are
Lyonia ovalifolia, Rhododendron arboreum, etc. and lower storey comprises
Berberis aristata, Coriaria nepalensis, Desmodium elegans, Lonicera quin-
quelocularis, Prinsepia utilis, Rubus ellipticus, R. niveus, Sinarundinaria
falcata, Toona sinensis, etc.
(b) Moru oak forests (Quercus dilatata forests)
It occurs only confined to nallahs above ban and below temperate moist
deciduous forests at an altitude of nearly 2250 m. The upper storey com-
prises Acer caesium, Aesculus indica, Alnus nepalensis, Lyonia ovalifolia,
Rhododendron arboreum, Quercus dilatata, etc., and the lower storey com-
prises Berberis aristata, Clematis montana, Desmodium elegans, Ilex dipy-
rena, Lonicera quinquelocularis, Parthenocissus semicordata, Rhamnus
virgatus, Rubus niveus, Sinarundinaria falcata, Viburnum cotinifolium, etc.
(c) Western mixed forests
This type of forest is found in Khati and Jatoli with altitudes from 2400
to 3000 m. The upper storey comprises Acer sp., Aesculus indica, Abies
spectabilis, Cedrus deodara, Juglans regia, Rhododendron arboreum,
R. campanulatum, Taxus baccata, Quercus dilatata, Q. leucotrichophora,
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 169
etc., and the lower storey comprises Adiantum venustum, Bromus unioloi-
des, Impatiens spp., Lonicera quinquelocularis, Oplismenus undulatifolius,
Strobilanthes atropurpureus, Valeriana jatamansi, Viburnum spp., Viola
pilosa, etc.
(d) Moist temperate deciduous forests
These forests occupy the cool and moist areas within the temperate region
between 2100 to 2750 m. Such forest occupy in between Jatoli and Kathalia.
The upper storey comprises Abies spectabilis, Aesculus indica, Betula alnoi-
des, Juglans regia, Quercus semicarpifolia, Rhododendron campanulatum,
Taxus baccata, etc., and the lower storey comprises Adiantum venustum,
Berberis sp., Bromus unioloides, Habenaria sp., Impatiens spp., Indigofera
cassioides, Lonicera quinquelocularis, Oplismenus undulatifolius, Pyrus
sp., Rosa macrophylla, R. sericea, Rubus niveus, Valeriana sp., Viburnum
grandiflorum, Viola pilosa, etc.
(e) Montane bamboo brakes
The occurrences of this type of ringals are in small patches found below
the Quercus leucotrichophora forests and near nallahs or running streams,
i.e. Loharkhet, Jatoli, Kathalia. It is confined between an altitude of 1500 m
and 2700 m. On higher altitudes, clumps of Gol ringal (Sinarundinaria fal-
cata) are replaced by deo-ringals (Thamnocalamus spathiflorus). Abies
spectabilis and Quercus semicarpifolia are common along with some high-
level mixed deciduous species are also found.
3. Himalayan dry-temperate forests
This type is found in extremely small strips on the river bank in between Jatoli
and Kathalia, where it has colonized in association with Hippophae salicifolia
and Populus ciliata.
4. Sub-alpine forests
(a) West Himalayan sub-alpine high-level birch forests
It occupies large tracts above 2000 m. The forests are dense and the trees
are short boled and ill formed. The upper storey comprises mainly Betula
utilis, Berberis spp., Lonicera angustifolia, Quercus semicarpifolia,
Rhododendron campanulatum, Rosa sericea, Salix acmophylla, Sorbaria
tomentosa, Viburnum grandiflorum, etc., and the lower storey comprises
Aconitum spp., Anemone obtusiloba, Allium sp., Corydalis sp., Impatiens
spp., Senecio sp., Pedicularis spp., Polygonum vaccinifolium, Potentilla sp.,
Saxifraga sp., Sibbaldia sp., Salvia sp., Thalictrum sp., Viola pilosa, etc.
(b) Sub-alpine pastures
This type of pastures occurs in fragmented patches near Jatoli, Kathalia,
Baloni top, Devikund, Sukhram cave, etc. It comprises shrubs, such as
Cotoneaster microphyllus, Juniperus sp., Rhododendron campanulatum,
Rosa sericea, Sorbaria tomentosa, Viburnum grandiflorum, etc., and herbs,
viz., Agropyron sp., Brachypodium sp., Bromus spp., Carex spp., Festuca
sp., Juncus spp., Poa spp., etc.
170 R. Manikandan et al.
3 Results
3.1 Ethno-Medicinal Plants
The following some wild ethno-medicinal plants have been reported from the valley
are Aconitum heterophyllum, A. violaceum, Anaphalis contorta, A. triplinervis,
A. virgata, Androsace sarmentosa, Anemone vitifolia, Angelica glauca, Aster albe-
scens, Cassiope fastigiata, Chaerophyllum villosum, Corydalis cashmeriana,
C. meifolia, Cotoneaster duthieana, Cynoglossum nervosum, C. wallichii, C. zeyl-
anicum, Dactylorhiza hatagirea, Delphinium brunonianum, Epilobium angustifo-
lium, Fritillaria roylei, Galium asperuloides, Gaultheria trichophylla, Gentiana
albicalyx, G. stipitata, Gentianella pedunculata, Gnaphalium hypoleucum,
Impatiens sulcata, Jurinea dolomiaea, Malaxis muscifera, Meconopsis aculeata,
Nardostachys grandiflora, Nepeta eriostachya, Oxyria digyna, Pedicularis hoff-
meisteri, P. punctata, Picrorhiza kurrooa, Polygonum cognatum, P. molliaeforme,
P. polystachyum, Potentilla atrosanguinea, P. cuneata, Primula elliptica, P. macro-
phylla, Ranunculus hirtellus, Rheum webbianum, Rhododendron anthopogon,
172 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Eriocapitella vitifolia Herb 2680 Paste of roots and leaves are
(Buch.-Ham. ex DC.) applied externally on ringworm
Nakai
Oxygraphis endlicheri Herb 3400
(Walp.) Bennet & Sum.
Chandra
Ranunculus diffusus DC. Herb 2000 Leaves and stems used in
rheumatism
Ranunculus hirtellus Herb 1900 Plant paste externally used on
Royle wounds
Thalictrum alpinum L. Herb 2000 Plant is used for heat-clearing and
detoxification/
Thalictrum foliolosum Herb 2100 Root is a tonic for dyspepsia, peptic
DC. ulcers, indigestion, fevers,
toothache, haemorrhoids and
ophthalmia. Leaf juice is applied to
boils and pimples
Trollius acaulis Lindl. Herb 3000 Plant is used to treat respiratory
tract infections, pharyngitis,
tonsillitis, bronchitis
Paeoniaceae
Paeonia emodi Royle Herb 2000 Leaves used against urinary
infections
Schisandraceae
Schisandra grandiflora Climber 2200 Fruits edible
(Wall.) Hook. f. &
Thomson
Menispermaceae
Cissampelos pariera L. Climber 2000 Plant used in the treatment of ulcer,
wound, rheumatism, fever, asthma,
cholera, diarrhoea, inflammation,
snake bite, malaria, rabies
Cocculus laurifolius DC. Shrub 2000 Plant cures rheumatic pain,
epilepsy, hypertension, abdominal
pain, headache, and scalp wounds
Berberidaceae
Berberis aristata DC. Shrub 2600 Plant is used against opthalmia, LC
fever. It is a blood purifier
Berberis asiatica Roxb. Shrub 2200 Plant is used in treating ulcers,
ex DC. urethral discharges, ophthalmia,
jaundice and fever
Berberis chitria Shrub 2700 Plant cures jaundice, splenomegaly,
Buch.-Ham. ex Lindl. leprosy, rheumatism, fever, snake
bite
(continued)
174 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Berberis jaeschkeana Shrub 2650 Roots and stems are used to make
C.K.Schneid. an eyewash to treat a range of eye
problems. Fruits edible
Berberis kumaonensis Shrub 2650 Plant can act as a good blood
C.K.Schneid. purifier and treat jaundice
Berberis napaulensis Tree 2000– Plant treats dysentery. The bark
(DC.) Spreng. 2900 decoction is used as eye drops to
treat eye inflammation
Papaveraceae
Papaver guilelmi- Herb 3200 Extract of petals is used to cure
waldemarii (Klotzsch) fever
Christenh. & Byng
Lardizabalaceae
Stauntonia latifolia Climber 3000 Fruit pulp is used to cure dyspepsia
(Wall.) R.Br. ex Wall. and flatulence
Fumariaceae
Corydalis cashmeriana Herb 2900 Root extracts applied externally to
Royle reduce body swelling and
inflammation
Corydalis cornuta Royle Herb 2850 Leaf juice mixed with equal
amount of water is taken orally to
cure bone fractures, torn ligaments
Corydalis diphylla Wall. Herb 1900 Plant is used for weight loss and
body care
Corydalis govaniana Herb 2800 Plant extract is used in eye diseases
Wall.
Corydalis meifolia Wall. Herb 3150 Root extract cures fever
Corydalis thyrsiflora Herb 3250 Plant cures stomach problems and
Prain. emotional problems
Fumaria indica Herb 2150 Plant is used in diarrhoea, fever,
(Hausskn.) Pugsley influenza and liver diseases
Brassicaceae
Arabis pterosperma Herb 2700
Edgew.
Arcyosperma Herb 3050
primulifolium (Thomson)
O.E. Schulz
Capsella bursa-pastoris Herb 1600 Plant is used as vegetable and in the LC
(L.) Medik. treatment of eye diseases and
dysentery
Cardamine flexuosa With. Herb 2850 Leaves edible. Leaves and seeds
used to treat skin infections
Cardamine impatiens L. Herb 2200 Plant is antirheumatic, diuretic and
stimulant. Leaves and young shoots
are used as vegetable
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 175
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Descurainia sophia (L.) Herb 2200– Plant is antipyretic, antipruritic,
Webb ex Prantl 4000 purgative, helminthic, digestive
tonic, cardiotonic, astringent
Nasturtium officinale Herb 3000 Plant cures urinary tract infections, LC
W.T.Aiton swollen airways, cough, bronchitis,
and mild muscular pain
Thlaspi arvense L. Herb 2000 Plant is anti-inflammatory and
febrifuge, used in the treatment of
pus in the lungs, renal
inflammation, appendicitis, seminal
and vaginal discharges
Violaceae
Viola betonicifolia var. Herb 2100 Plant is diaphoretic, purgative,
nepalensis (Ging.) Back. astringent, anticancer, antipyretic,
and to treat nervous disorders,
cough, skin diseases, sinusitis,
kidney diseases, pneumonia, and
bronchitis
Viola biflora L. Herb 2800 Plant used in stomach and intestinal
swellings, digestion problems, gas,
heartburn, gall bladder disorders,
and loss of appetite
Viola hamiltoniana Herb 2500
D. Don
Viola pilosa Blume Herb 2000 Flower extract is used to cure
jaundice
Caryophyllaceae
Acanthophyllum Herb 2850
cerastioides (D. Don)
Madhani & Zarre
Arenaria neelgherrensis Herb 2050 Plant is used to treat urinary tract
Wight & Arn. disorders, kidney and bladder
stones
Eremogone festucoides Herb 2050 Taken as tonic to increase lactation
(Benth.) Pusalkar &
D.K. Singh
Sagina saginoides (L.) Herb 2700 Plant is used to treat food poisoning
H. Karst. and diarrheoa
Silene setaesperma Herb 2550
Majumdar
Silene vulgaris (Moench) Herb 2500 The extract of young shoot is used LC
Garcke to treat bronchitis and asthma
Stellaria decumbens Herb 2700
Edgew.
(continued)
176 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Stellaria media (L.) Vill. Herb 2100 Plant is used to relieve itching and LC
psoriasis and applied as a plaster
for broken bones and swellings
Stellaria monosperma Herb 2200 Plant parts can be used as vegetable
var. paniculata (Edgew.)
Majumdar
Tamaricaceae
Myricaria germanica (L.) Shrub 1800– Wood is used as fuel. Decoction of
Desv. 4000 the bark is aperient, used to treat
jaundice
Hypericaceae
Hypericum choisyanum Shrub 2050 Plant extracts an essential oil
Wall. ex N.Robson
Hypericum elodeoides Herb 2400 Plant is used in the treatment for
Choisy anxiety, depression, cuts, and burns
Hypericum hookerianum Shrub 2500 Plant is a potential wound-healing
Wight & Arn. agent
Hypericum japonicum Herb 2500 Plant is used in the treatment of
Thunb. asthma and dysentery, hepatitis,
appendicitis, boils and abscesses
Hypericum oblongifolium Shrub 2100 Plant is used against hepatitis,
Choisy gastric ulcers, and bee stings
Hypericum uralum Herb 2200
Buch.-Ham. ex D. Don
Theaceae
Eurya acuminata DC. Tree 2150 Leaf decoction is used to cure
cholera, diarrhoea and other
stomach diseases
Geraniaceae
Geranium collinum Herb 2940 Plant cures rheumatism, gout,
Stephan ex Willd. dysentery, external and internal
bleeding, eczema, scabies
Geranium nepalense Herb 2850 Plant used in the treatment of
Sweet. nervous disorders, limb numbness,
rheumatism, renal diseases
Geranium ocellatum Herb 2200 Decoction of whole plant is used to
Jacquem. ex Cambess. control dysentery in goats and
sheep
Geranium robertianum L. Herb 2740 Plant used to treat diarrheoa, to
improve functioning of the liver
and gallbladder, for toothache and
nosebleeds
Geranium wallichianum Herb 2950 Plant used in the treatment of LC
D. Don ex Sweet. peptic ulcers, toothache
Oxalidaceaea
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 177
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Oxalis corniculata L. Herb 2000 Plant is used in influenza, fever,
urinary tract infections, enteritis,
diarrhoea, traumatic injuries,
sprains and poisonous snake bites
Oxalis dehradunensis Herb 2800 Plant used in the treatment of fever,
Raizada pain, inflammation and wound
healing
Balsaminaceae
Impatiens devendrae Herb 2700
Pusalkar
Impatiens edgeworthii Herb 2250 Plant is used for gonorrhea, burns
Hook.f.
Impatiens racemosa DC. Herb 2150 Leaves and roots paste mixed with
mustard oil applied for rheumatic
pains
Impatiens scabrida DC. Herb 2000
Impatiens sulcata Wall. Herb 2750 Paste of flower buds applied on
pimples
Impatiens thomsonii Herb 2100
Hook. f.
Rutaceae
Boenninghausenia Herb 2100 Leaf paste is applied to cuts and
albiflora (Hook.) Rchb. wounds. Crushed leaves can be
ex Meisn. placed in the nostrils to treat
malaria
Skimmia anquetilia Shrub 2730 Leaves are used in religious
N.P.Taylor & Airy Shaw ceremonies and also as incense
‘dhup’
Skimmia laureola (DC.) Shrub 2400 Leaves are used in the treatment of
Decne. smallpox
Zanthoxylum oxyphyllum Shrub 2065 Bark is used in the treatment of
Edgew. rheumatism. A paste made from
immature fruits is held between the
teeth for about 10 minutes to
relieve toothache
Aquifoliaceae
Ilex dipyrena Wall. Tree 2200 The wood is used for fuel LC
Celastraceae
Euonymus echinatus Shrub 2700 Plant cures respiratory problems, LC
Wall. asthma. The root extract is effective
in treating whooping cough,
malarial fever and renal colic
Euonymus tingens Wall. Tree 2230 The juice of the bark is used in the
treatment of eye diseases, chronic
constipation and dyspepsia
(continued)
178 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Rhamnaceae
Rhamnus procumbens Shrub 2000 Plant is used as cardiac stimulant,
Edgew. CNS depressant and analgesic
Rhamnus purpurea Shrub 2700–
Edgew. 3000
Rhamnus virgata Roxb. Shrub 1950 The fruit is used in the treatment of LC
spleen disorders
Vitaceae
Ampelocissus latifolia Climber 2050 Plant used in the treatment of bone
(Roxb.) Planch. fractures, dysentery, leucorrhoea,
dental problems, stomach pain,
gout, tuberculosis, dyspepsia,
indigestion, body weakness
Parthenocissus Climber 2100 The poultice of the roots is used to
semicordata (Wall.) set dislocated bones
Planch.
Parthenocissus Climber 1900– Fruits edible
semicordata var. roylei 3000
(King ex R.Parker)
Nazim. & Qaiser
Tetrastigma obtectum Climber 2100 Plant possess anticancer, anti-
(Wall. ex M.A. Lawson) inflammatory, antimicrobial
Planch. ex Franch. activities
Hippocastanaceae
Aesculus indica (Wall. ex Tree 2035 Plant is used in skin diseases, LC
Cambess.) Hook.f. rheumatism, headache
Aceraceae
Acer acuminatum Wall. Tree 2450 Leaves are used as a tea substitute LC
ex D. Don
Acer caesium Wall. ex Tree 2400 Plant is anti-rheumatic, purgative, LC
Brandis anti-inflammatory, emetic
Acer cappadocicum Gled. Tree 2350 Wood is used for construction, LC
ploughs, bedsteads and poles to
carry loads
Acer oblongum Wall. ex Tree 2300 Wood is used for agricultural LC
DC. implements, minor construction,
cups
Acer villosum Wall. Tree 2800 The wood is used for agricultural LC
implements. Leaves used as fodder
Staphyleaceae
Staphylea emodi Wall. ex Shrub 2100 Plant is used for making walking
Brandis sticks
Sabiaceae
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 179
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Meliosma dilleniifolia Tree 2200 Good fodder
(Wall. ex Wight & Arn.)
Walp.
Anacardiaceae
Pistacia chinensis subsp. Tree 2100 Plant used in the treatment of
integerrima (J.L. Stewart) asthma, diarrheoa, diabetes, liver
Rech.f. diseases, fever, pain and
inflammation
Brucea javanica (L.) Tree 2100 Leaves are used against enlarged LC
Merr. spleen, scurf, ringworm, boils and
centipede bites
Toxicodendron wallichii Tree 2100 Leaf juice is a corrosive vesicant LC
(Hook.f.) Kuntze
Papilionaceae
Astragalus chlorostachys Shrub 2950 Root juice taken as a tonic for
Lindl. longevity and strength
Astragalus himalayanus Herb 2750 Powdered flowers and seeds are
Klotzsch given in strangury
Biancaea decapetala Climber 2050 Roots, stems and pods act as pain LC
(Roth) O.Deg. reliever. Also the paste is applied to
burns
Campylotropis speciosa Shrub 2300
subsp. eriocarpa
(Schindl.) Iokawa & H.
Ohashi
Dumasia villosa DC. Climber 2500 Seeds contain essential oil LC
Hylodesmum podocarpum Herb 2100 Plant is used to cure fever, malaria,
subsp. oxyphyllum (DC.) coughs and bleeding wounds
H. Ohashi & R.R. Mill
Leptodesmia microphylla Shrub 1900
(Thunb.) H. Ohashi &
K. Ohashi
Lespedeza gerardiana Herb 2560 Seeds crushed and used for body
Wall. ex Maxim. massage
lndigofera cylindrica Shrub 2500 Plant cures cough and cold
Grah. & Baker.
Lndigofera heterantha Shrub 2100 Flowers are boiled and pickled.
Wall. ex Brandis Branches are used to make basket,
twig bridges
Lotus corniculatus L. Herb 2050 Plant is used against skin LC
inflammation
Ototropis multiflora (DC.) Shrub 2050 Branches and flowers are used for
H. Ohashi & K. Ohashi reducing fever by inducing sweat
Parochetus communis Creeper 2000– LC
Buch.-Ham. ex D.Don 3000
(continued)
180 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Piptanthus nepalensis Shrub 2150
(Hook.) Sweet
Senna occidentalis (L.) Shrub 1700 Plant is diuretic, laxative, blood LC
Link. purifier, antibacterial, and
antifungal. It is used in the
treatment of haemorrhoids, gout,
rheumatism, diabetes, whooping
cough
Senna tora (L.) Roxb. Shrub 1400 Young shoots and leaves are cooked
and eaten. The powdered and
fermented leaves are used as a
condiment
Shuteria involucrata Climber 2300 A paste of the plant is used to set
(Wall.) Wight & Arn. ex dislocated bones
Walp.
Smithia ciliata Royle Herb 1300
Sunhangia elegans (DC.) Shrub 2600 The juice of the bark is used in the
H. Ohashi & K. Ohashi treatment of peptic ulcers
Tateishia concinna (DC.) Shrub 2100
H. Ohashi & K. Ohashi
Trifolium repens L. Herb 2700 A tincture of the leaves is applied LC
to gout. An infusion of the flowers
is used as eyewash
Trigonella gracilis Benth. Herb 2200– A paste made from the plant is
3400 applied topically to treat skin
diseases
Vicia rigidula Royle Climber 1800
Zornia gibbosa Span. Herb 1200 Plant is anti-inflammatory and
astringent; is used in the treatment
of inflammations and dysentery
Rosaceae
Agrimonia pilosa Ledeb. Herb 2000 Plant is used for abdominal pain, LC
sore throat, headache, heat stroke,
boils, eczema and taeniasis
Agrimonia pilosa Ledeb. Herb 1000– Leaf decoction is effective against
subsp. japonica (Miq.) 3000 liver problems
Hara var. japonica (Miq.)
Nakai
Argentina lineata (Trevir.) Herb 2800
Soják
Argentina micropetala Herb 3100 Stolons and stems are astringent
(D.Don) Soják
Argentina microphylla Herb 3500
(D.Don) Soják
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 181
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Argentina polyphylla Herb 3200
(Wall. ex Lehm.) Soják
Aruncus dioicus (Walter) Herb 3050 A tea made from the roots is used
Fernald to allay bleeding after child birth, to
reduce profuse urination and to
treat stomach pains, diarrhoea,
gonorrhoea, fevers and internal
bleeding
Cotoneaster acuminatus Shrub 3000
Lindl.
Cotoneaster adpressus Shrub 2550 Young fruits taken to cure diarrhoea
Bois
Cotoneaster affinis Lindl. Shrub 3000
Cotoneaster microphyllus Shrub 2650 Stems are astringent. Leaves used
Wall. ex Lindl. as incense. Fruits edible
Dasiphora fruticosa (L.) Herb 2340
Rydb.
Filipendula vestita (Wall. Herb 3030
ex G.Don) Maxim.
Fragaria daltoniana Herb 3000 The juice of root is used in the
J.Gay treatment of fever. Fruits edible
Fragaria nubicola (Lindl. Herb 2750 The unripe fruit is chewed to treat
ex Hook.f.) Lacaita blemishes on tongue. Fruits edible
Geum elatum Wall. ex Herb 3000 Plant used in diarrheoa,
G.Don inflammation, haemorrhoids and
stomach problems
Geum urbanum L. Herb 2340 Plant is used to treat problems
affecting the mouth, throat and
gastrointestinal tract. It reduces
irritation of the stomach and gut
Griffitharia vestita (Wall. Tree 1960
ex G.Don) Rushforth
Potentilla argyrophylla Herb 2650
Wall. ex. Lehm.
Potentilla atrosanguinea Herb 3060 Crushed flowers applied to reduce
G.Lodd. ex D.Don the pain caused by insect stings
Potentilla crantzii Herb 1065
(Crantz) Beck ex Fritsch
Potentilla eriocarpa Wall. Herb 3300
ex Lehm.
Potentilla fulgens Wall. ex Shrub 1800 Root peel extract is used to get rid
Sims of intestinal parasitic infections
Potentilla indica Herb 2400
(Andrews) Th.Wolf
(continued)
182 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Potentilla nepalensis Herb 2100 Root is depurative. Ashes mixed
Hook. with oil and applied to burns
Potentilla purpurea Herb 3040 Plant used in the treatment of
(Royle) Hook.f. inflammations, wounds
Potentilla sundaica Herb 1400 Decoction of the plant is used in the
(Blume) W. Theob. treatment of colds, influenza, sore
throat
Prunus cornuta (Wall. ex Tree 2700 Fruits and seeds are edible
Royle) Steud.
Prunus nepalensis Ser. Tree 2500 A green dye can be obtained from
the leaves
Rosa brunonii Lindl. Climber 1700 Plant stimulates wound healing
Rosa macrophylla Lindl. Shrub 2200 Fruit paste is used against eye sight
issues
Rosa sericea Lindl. Shrub 2630 Plant cures uterine diseases. A
paste of the flowers is applied on
forehead to cure headache
Rubus ellipticus Sm. Shrub 2050 Plant juice can be used to treat LC
coughs, fevers, colic and sore throat
Rubus nepalensis Shrub 2500 Fruits edible
(Hook.f.) Kuntze
Rubus niveus Thunb. Shrub 2100 Fruits edible
Rubus paniculatus Sm. Climber 1750 A paste of the bark is used in the
treatment of scabies. Leaf paste is
used as a poultice on sprains
Rubus rosifolius Sm. Shrub 2200 A decoction of the root is used in
the treatment of diarrhoea. Leaf
paste is applied to itches
Sanguisorba diandra Herb 3000
(Hook.f.) Nordbong
Sibbaldia cuneata Edgew. Herb 3035
Sibbaldia cuneifolia Herb 3000
(Bertol.) Paule & Soják
Sorbus foliolosa (Wall.) Shrub 3000 LC
Spach
Spiraea bella Sims Shrub 3050
Spiraea canescens D.Don Shrub 1700 Plant is used to make walking LC
sticks
Saxifragaceae
Astilbe rivularis Shrub 2300 Plant is used for the treatment of
Buch.-Ham. ex D.Don ulcer, bleeding during child birth,
inflammation, body ache, diarrhoea
and dysentery
Bergenia ciliata (Haw.) Herb 2100 Plant treats urinary troubles. Leaf LC
Sternb. juice used as ear drops
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 183
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Bergenia pacumbis Herb 1500 Plant is a tonic in the treatment of LC
(Buch.-Ham. ex D.Don) fevers, diarrhoea and pulmonary
C.Y.Wu & J.T.Pan affections
Bergenia stracheyi Herb 3500
(Hook.f. & Thomson)
Engl.
Chrysosplenium tenellum Herb 2450
Hook.f. & Thomson
Chrysosplenium Herb 2840
trichospermum Edgew. ex
Hook.f. & Thomson
Micranthes pseudopallida Herb 2795
(Engl. & Irmsch.)
Losinsk.
Saxifraga asarifolia Herb 3200–
Sternb. 4600
Saxifraga brachypoda Herb 2960
D.Don
Saxifraga brunonis Wall Herb 3250
ex Ser.
Saxifraga diversifolia Herb 2400–
Wall. ex. DC. var. 4600
parnassifolia (D.Don)
Engl.
Saxifraga filicaulis Wall. Herb 2050
ex Ser.
Saxifraga flagellaris Herb 3000
Willd.
Saxifraga jacquemontiana Herb 4000 Plant extract is act as liver tonic
Decne.
Saxifraga parnassifolia Herb 3000
D.Don
Saxifraga sibirica L. Herb 3500
Saxifraga stenophylla Herb 2500 Root paste applied to cure
Royle toothache
Parnassiaceae
Parnassia nubicola Wall. Herb 3300 Plant extract is used for the
ex Royle treatment of food poisoning.
Rootstocks are used externally in
snakebites
Hydrangeaceae
Deutzia compacta Craib Shrub 2500
Deutzia staminea R.Br. ex Shrub 2250 Plant is diuretic
Wall.
(continued)
184 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Hydranzea anomala Shrub 2100 A boiled concoction of the leaves is
D.Don used to make a syrup
Philadelphus tomentosus Shrub 2000
Wall. ex G.Don
Grossulariaceae
Ribes glaciale Wall. Shrub 3000 Fruits edible
Ribes griffithii Hook.f. & Shrub 2500 Fruits edible
Thomson
Ribes himalense Royle ex Shrub 2000– The Leaf juice is used in the
Decne. 3300 treatment of diarrhoea and
dysentery
Crassulaceae
Crassula campestris Herb 3000 LC
(Eckl. & Zeyh.) Endl.
Hylotelephium ewersii Herb 3250
(Ledeb.) H.Ohba
Rhodiola bupleuroides Herb 3500 Juice of rhizomes given to cure
(Wall. ex Hook.f. & whooping cough
Thomson) S.H. Fu
Rhodiola Herb 2500
chrysanthemifolia
(H.Lév.) S.H. Fu
Rhodiola heterodonta Herb 3500 Shoots are used as a remedy against
(Hook. f. & Thomson) cold, cough and lung infections
Boriss.
Rhodiola quadrifida Herb 2860 Plant act as a tonic, antidepressant
(Pall.) Fisch. & C.A. Mey and anti-inflammatory drug
Rhodiola sinuata (Royle Herb 3000 Plant is used for increasing energy,
ex Edgew.) S.H. Fu endurance, strength, and mental
capacity
Rhodiola wallichiana Herb 3050 Plant is used in the treatment of
(Hook.) S.H. Fu cardiovascular diseases, diarrheoa,
hysteria, hernias, headache
Sedum multicaule Wall. Herb 3032 Plant is emollient and vulnerary
ex Lindl.
Sedum oreades (Decne.) Herb 3600 Leaf decoction is taken to cure
Raym.-Hamet cough and cold
Tillaea pentandra Royle Herb 1900– Root decoction is laxative and used
2500 to treat fevers
Melastomaceae
Osbeckia stellata Herb 2300
Buch.-Ham. ex D.Don
Onagraceae
Circaea alpina L. Herb 2100 Plant used to treat tumours, fresh
cuts or wounds
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 185
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Epilobium amurense Herb 3200
Hausskn.
Epilobium angustifolium Herb 2450 Root paste is applied for wounds LC
L.
Epilobium latifolium L. Herb 2780 Plant is used in the treatment of LC
fevers, inflammations, itching
pimples
Epilobium palustre L. Herb 4500 LC
Epilobium roseum Herb 1500–
(Schreb.) Schreb. 2000
Epilobium royleanum Herb 3300
Hausskn.
Cucurbitaceae
Solena amplexicaulis Climber 2600 Plant is used in anorexia, digestive
(Lam.) Gandhi problems, flatulence, asthma,
gonorrhoea, inflammation
Begoniaceae
Begonia picta Sm. Herb 2900 Plant juice is taken to relieve
headache, peptic ulcer. The paste is
applied to stop bleeding from cuts
and wounds
Apiaceae
Acronema tenerum (DC.) Herb 2800
Edgew.
Angelica glauca Edgew. Herb 3350 Root powder is mixed with milk EN
given in empty stomach to cure
severe stomach pain
Bupleurum hamiltonii Herb 3000 Root is used to regulate the
N.P. Balakr. metabolism, for the treatment of
fever, pain and inflammation
Bupleurum lanceolatum Herb 1400
Wall. ex DC.
Bupleurum longicaule Herb 3000 Plant is used against chronic
Wall ex DC. hepatitis, nephrotic syndrome and
autoimmune diseases
Centella asiatica (L.) Herb 600 Plant is a brain tonic, to improve LN
Urb. memory power also is used to treat
leprosy, lupus, varicose ulcers,
eczema, psoriasis, diarrhoea, fever,
amenorrhoea
Chaerophyllum reflexum Herb 3200
var. acuminatum (Lindl.)
Hedge & Lamond
Chaerophyllum villosum Herb 3000 Cooked roots are nutritious
Wall. ex DC.
(continued)
186 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Cortia candollei (DC.) Herb 2650
Leute
Heracleum candicans Herb 1800– Root is used for the treatment of
Wall.ex DC. 5000 leucoderma. Decoction of root and
stem cures fever. Root paste is
applied on snake bite
Hydrocotyle javanica Herb 2800 Plant is used to cure sore throats LC
Thunb. and lungs. Leaf juice is used as eye
drops to cure eye infection. Leaf
paste is used in dressing of wounds
to reduce swelling
Hydrocotyle Creeper 600– Plant cures fevers, wounds and LC
sibthorpioides Lam. 2500 boils
Hymenidium brunonis Herb 4000
(DC.) Lindl.
Hymenolaena candollei Herb 3800
DC.
Ligusticopsis wallichiana Herb 2900– Leaf extract applied on cut and
(DC.) Pimenov & 3100 wounds
Kljuykov
Ligusticum striatum DC. Herb 1500– Plant is used in the treatment of
3500 migraine, cardiovascular and
cerebrovascular diseases
Pimpinella diversifolia Herb 2300 Leaves used to treat stomach ache
DC.
Sanicula elata Buch.- Herb 2100
Ham. ex D.Don
Selinum vaginatum Herb 3000
(Edgew.) C.B.Clarke
Trachydium roylei Lindl. Herb 2990 Plant contains an essential oil that
is used in the treatment of
inflammation
Vicatia coniifolia DC. Herb 2800
Araliaceae
Hedera nepalensis Climber 2000 Plant used in the treatment of
K. Koch. diabetes
Cornaceae
Cornus macrophylla Wall. Tree 2400 Plant cures malaria, allergy, LC
inflammation, diabetes, cancer
Caprifoliaceae
Leycesteria formosa Wall. Shrub 2550 The hollow stems can be made into
whistles and flutes
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 187
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Lonicera angustifolia var. Shrub 2700
myrtillus (Hook.f. &
Thomson) Q.E.Yang,
Landrein, Borosova &
Osborne
Lonicera hispida Pall. ex Shrub 2900–
Schult. 4500
Lonicera japonica Thunb. Climber 1000 Plant is used to cure digestive
disorders, pain and swelling of
small intestine
Lonicera purpurascens Shrub 1500
(Jacquem. ex Decne.)
Walp.
Lonicera Shrub 2100 Powdered leaves are used for
quinquelocularis Hardw. healing wounds. Plant material is
used as fuel
Viburnum cotinifolium Shrub 2850 Leaf extract is used for the
D.Don treatment of menorrhagia. Fruits
edible
Viburnum erubescens Shrub 1500– The juice of the roots is used in the
Wall. ex DC. 2700 treatment of coughs. Fruits edible
Viburnum grandiflorum Shrub 2550 The fruit is used in the treatment of
Wall. ex DC. typhoid and whooping cough
Viburnum mullaha Shrub 2200 Juice of fruit is used to treat LC
Buch.-Ham.ex D.Don indigestion
Viburnum nervosum Shrub 2700 Fruits edible
D.Don
Rubiaceae
Galium acutum Edgew. Herb 3000
Galium aparine L. Herb 2100 Plant is used to treat painful
urination, infected lymph nodes,
psoriasis
Galium asperifolium Herb 2800
Wall.
Galium asperuloides Herb 2800 Plant paste applied on skin itching
Edgew.
Galium triflorum Michx. Herb 1500– LC
3000
Himalrandia tetrasperma Shrub 2050 Plant possess anti-inflammatory,
(Wall. ex Roxb.) anti-bacterial. Antifungal, anti-viral
T. Yamaz. and anti-malarial activities
Leptodermis lanceolata Herb 2000 Bark paste is applied on forehead to
Wall. treat migraine
Rubia cordifolia L. Herb 2600– Plant detoxifies the blood
4500
(continued)
188 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Spermadictyon Shrub 2050 Root extracts are used in the
suaveolens Roxb. treatment of diabetes, rheumatoid
arthritis and bloody dysentery
Valerianaceae
Nardostachys jatamansi Herb 3200 Plant extracts used in heart-pain, CR
(D.Don) DC. fits and rubbed in muscle fracture
to relieve pain also regulate
urination, menstruation and
digestion
Valeriana hardwickei Herb 2860 Plant helps to improve sleep quality
Wall. and reduce blood pressure
Valeriana himalayana Herb 3250 Plant helps reduce sleeping
Grubov disorders, especially insomnia,
anxiety, stress, etc
Valeriana jatamansi Jones Herb 2400 Plants are useful in epilepsy,
ex Roxb. hysteria, hypochondriasis, nervous
unrest, and skin diseases
Dipsacaceae
Dipsacus inermis Wall. Herb 2600 The root is used to promote bone
growth and to correct bone
fractures
Triplostegia glandulifera Herb 3000 Plant cures kidney problems
Wall. ex DC.
Morinaceae
Morina longifolia Wall. Herb 3000 Plant is used in the treatment of
ex DC. stomach disorders, such as
indigestion, vomiting and nausea
Asteraceae
Adenocaulon himalaicum Herb 2750 Plant is used in treating abscesses,
Edgew. haemorrhage
Ageratum conyzoides L. Herb 2500 Plant is used to treat constipation, LC
infective hepatitis, eczema,
epilepsy, fresh wounds, dizziness,
diarrhoea, dysentery, sore eyes,
fever, headache, intestinal worms,
filariasis, vomiting and nausea,
wounds and cuts
Ainsliaea aptera DC. Herb 2150 Roots paste is used to relieve
stomach ache, constipation, fever
Ainsliaea latifolia Herb 2450 Plant used in the treatment of
(D.Don) Sch.-Bip. rheumatism, traumatic injuries,
edema, stomach ache, and anorexia
Allardia glabra Decne. Herb 3700 Plant paste applied to heal wounds
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 189
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Anaphalis busua Herb 2450 Plant is used for diarrhoea,
(Buch.-Ham. ex D.Don.) dysentery and pulmonary
affections. A poultice of flowers or
the whole plant is applied to burns,
sores, ulcers, bruises, swellings and
rheumatic joints
Anaphalis contorta Herb 2300 Smoke of the plant is used as an
(D.Don.) Hook f. insect repellent and paste of young
flower heads applied on pimples
Anaphalis margaritacea Herb 3000 Plant is a good remedy for
(L.) Benth. & Hook f. diarrhoea, dysentery and pulmonary
affections
Anaphalis royleana DC. Herb 2750 Plant is antibacterial, anti-
inflammatory, antirheumatic,
antiseptic, antiviral, diaphoretic,
expectorate, sedative
Anaphalis triplinervis Herb 2630 Paste of flower heads applied on
(Sims.) C.B Clarke cuts, wounds and boils
Anaphalis virgata Herb 2600 Smoke of the plant used as an
Thomson. ex C.B.Clarke insect repellent and paste of whole
plant applied on wounds
Artemisia capillaris Herb 100– Plant cures liver diseases
Thunb. 2700
Artemisia nilagirica Herb 2000 Seed oil is extracted and massaged
(C.B.Clarke) Pamp. on swollen joints in case of
arthritis, back pain and bone
fractures
Artemisia roxburghiana Herb 2600 Plant used against colic, diarrheoa,
Besser constipation, cramps, indigestion,
worm infestations, vomiting
Artemisia vestita Wall. ex Shrub 2100 The plant is used in anti-
Besser inflammatory and antifebrile
medicines
Aster albescens (DC.) Herb 2700 Plant extracts cure tooth ache
Wall. ex Hand.-Mazz.
Aster diplostephioides Herb 2800 Flowers are used in the treatment of
(DC.) Benth. ex infectious fevers, influenza, nose
C.B.Clarke bleeds, poisoning, sores
Baccharoides Herb 1700 Seeds used in the treatment of skin
anthelmintica (L.) diseases, scorpion stings
Moench
Bidens pilosa L. Herb 2100 Roots are used to treat constipation
and malaria, toothache
(continued)
190 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Cirsium arvense (L.) Herb 2500 The root is tonic, diuretic,
Scop. astringent, antiphlogistic and
hepatic. It is chewed to cure
toothache. Root decoction is used
to treat worms in children
Cirsium verutum Herb 2200 Root extract is used to relieve
(D.Don.) Spreng. fevers. Root paste is used in the
treatment of stomach disorders. The
fresh root is chewed to treat
nosebleeds and throat pain
Cirsium wallichii DC. Herb 2300 Plant is used to treat fever and nose
bleeding
Cordiofontis peduncularis Herb 2750 Root decoction applied to wounds
(Wall. ex Nees)
G.L.Nesom
Cremanthodium dacaisnei Herb 3000
C.B.Clarke
Dolomiaea macrocephala Herb 3200 Incense of plant used to cure
DC. ex Royle breathing problems
Doronicum kamaonense Herb 2400
(DC.) Alv. Fern
Dubyaea hispida DC. Herb 2440
Duhaldea nervosa (Wall. Herb 1200–
ex DC.) Anderb. 2000
Erigeron alpinus L. Herb 3800 Plant is used for cough, cold,
nausea, gastric and rheumatism
Erigeron multiradiatus Herb 3300 Plant is used in inflammatory
(Lindl. ex DC.) Benth. & diseases
Hook.f.
Eschenbachia japonica Herb 2100
(Thunb.) J.Kost.
Eschenbachia stricta Herb 1550
(Willd.) Raizada
Galinsoga parviflora Cav. Herb 2200 Plant used to treat eczema, wounds
Gamochaeta pensylvanica Herb 4700 Plant is used against diabetes, high
(Willd.) Cabrera blood pressure, stomach ulcers,
diarrheoa, gut infections
Hippolytia longifolia Herb 3600
(Rech.f.) C.Shih
Jacobaea analoga (DC.) Herb 3000–
Veldkamp 3500
Jurinea albescens (DC.) Herb 3050
N.Garcia, Herrando &
Susanna
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 191
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Jurinea auriculata (DC.) Herb 3000
N.Garcia, Herrando &
Susanna
Leibnitzia nepalensis Herb 3200–
(Kunze) Kitam 4600
Leontopodium Herb 3700
himalayanum DC.
Ligularia amplexicaulis Herb 3400 Plant is used in the treatment of
DC. vomiting
Ligularia sibirica (L.) Herb 2750 Leaves cooked and eaten DD
Cass.
Melanoseris brunoniana Herb 2330
(Wall. ex DC.) N.Kilian
& Ze H.Wang
Melanoseris cyanea Herb 2400
(D.Don) Edgew.
Melanoseris macrorhiza Herb 3300 Juice of fresh rootstock is given to
(Royle) N.Kilian stomach pain
Melanoseris violifolia Herb 3000
(Decne.) N.Kilian
Mulgedium lacertianum Herb 3160
Wall. ex DC.
Myriactis wallichii Less. Herb 2730
Neobrachyactis anomala Herb 2900
(DC.) Brouillet
Oreoseris gossypina Herb 2300
(Royle) X.D.Xu &
V.A.Funk
Pentanema orientale Herb 2730
(Lam.) D.Gut.Larr.,
Santos-Vicente, Anderb.,
E.Rico & M.M.Mart.Ort.
Pseudognaphalium affine Herb 2100
(D.Don) Anderb.
Pseudognaphalium Herb 2100 Plant extract applied on cuts and
hypoleucum (DC.) wounds
Hilliard & B.L.Burtt
Saussurea Herb 3500
leontodontoides (DC.)
Sch.Bip.
Saussurea obvallata Herb 4100 Extract of petals applied for septic
(DC.) Sch.Bip. wounds
Saussurea roylei (DC.) Herb 3500 Plant is used in the treatment of
Sch.Bip. wounds Paste of the plant is used as
poultice for joint pains
(continued)
192 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Senecio graciliflorus DC. Herb 2500
Sigesbeckia orientalis L. Herb 2400 Plant used against arthritis,
rheumatism, and gout
Solidago virgaurea L. Herb 2500 Plant is used for gout, joint pain, LC
arthritis, eczema and other skin
diseases
Synotis alata (Wall.ex Herb 2400
DC.) C. Jeffrey &
Y.L. Chen
Synotis chenopodiifolia Herb 2660
(DC.) M.Tang, C.Ren &
Q.E.Yang
Synotis kunthiana (Wall. Herb 3050
ex DC.) Jeffrey &
Y.L.Chen
Synotis rufinervis (DC.) Herb 2400
C. Jeffrey & Y.L.Chen
Tagetes minuta L. Herb 2100 Plant used against digestive tract
problems including poor appetite,
gas, stomach pain, colic, intestinal
worms and dysentery
Taraxacum officinale Herb 4500 Roots extract cures headache LC
F.H.Wigg.
Tragopogon gracilis Herb 2300 Young shoots and fruits can be
D.Don eaten
Xanthium strumarium L. Herb 2000 Plant used in rhinitis, nasal
sinusitis, headache, gastric ulcer,
urticaria, rheumatism, arthritis and
microbial infections
Campanulaceae
Campanula argyrotricha Herb 3020
Wall.ex A.DC.
Campanula pallida Wall. Herb 2300
Codonopsis rotundifolia Climber 3000 Plant act as the protection against
Benth. side effects of radiation treatment,
heartburn
Codonopsis viridis Wall. Climber 1200–
3000
Cyananthus lobatus Wall. Herb 3000 Plant is used in the treatment of
ex Benth. papetic ulcer, serous disorders and
constipation
Cyananthus microphyllus Herb 3300
Edgew.
Lobelia pyramidalis Wall. Herb 1500– Plants are useful in treating asthma,
3000 bronchitis, and fever
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 193
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Ericaceae
Cassiope fastigiata Herb 3100 Incense of whole plant is inhaled to
(Wall.) D.Don ease respiration
Gaultheria Herb 2800 Fruits edible
nummularioides D.Don.
Gaultheria trichophylla Herb 3100 Plant used to given for cattle for
Royle lactation and immune disease
resistant
Lyonia ovalifolia (Wall.) Tree 1750 Young leaves and buds are used LC
Drude externally as an infusion to treat
skin diseases and external parasites
Rhododendron Shrub 3000 Decoction of young shoot given to
anthopogon D.Don cure fever
Rhododendron arboreum Tree 1500– Leaf paste is applied to the LC
Sm. 3300 forehead in the treatment of
headaches. The juice of the bark is
used in coughs, diarrhoea and
dysentery
Rhododendron barbatum Shrub 2400–
Wall ex G. Don 3700
Rhododendron Shrub 3600 Paste of flower buds applied on cuts
campanulatum D. Don.
Rhododendron lepidotum Shrub 2800 Leaf decoction is used against
Wall. ex D.Don colds, coughs, chronic bronchitis,
asthma and excessive mucus
formation in the nose or throat
Monotropaceae
Monotropastrum humile Herb 2500 Plant is used in the treatment of
(D.Don.) H.Hara cough
Primulaceae
Androsace geraniifolia Herb 2700–
Watt 3000
Androsace globifera Herb 2200
Duby
Androsace sarmentosa Herb 2300 Plant is used to cure tumours,
Wall. inflammations, serous fluid
disorders
Lysimachia lobelioides Herb 100–
Wall. 2000
Lysimachia prolifera Herb 2600
Klatt.
Primula denticulata Sm. Herb 3200 Raw flowers are used to make
salads
Primula elliptica Royle Herb 3200 Root paste applied for pimples and
to kill lice
(continued)
194 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Primula macrophylla Herb 3100 Plant extract is given to cure
D.Don urinary complaints
Primula munroi Lindl. Herb 3000–
4500
Primula stuartii Wall. Herb 3500
Myrsinaceae
Myrsine africana L. Shrub 2700 Plant cures skin allergies, boils
Symplocaceae
Symplocos Tree 1100 Plant is astringent, acrid,
cochinchinensis (Lour.) refrigerant, anti-inflammatory,
S. Moore depurative and febrifuge
Symplocos paniculata Tree 2400 Plant used in the treatment of
Miq. dysentery, bowel complaints,
inflammations, snake bites, vaginal
discharges and miscarriages
Symplocos ramosissima Tree 2200 LC
Wall. ex G.Don
Symplocos theifolia Tree 2000
D. Don
Oleaceae
Fraxinus micrantha Tree 2200 Plant is used against malaria and DD
Lingelsh. pneumonia
Jasminum dispermum Climber 2050 The roots and leaves are active
Wall. against ringworm and tapeworm
infections
Osmanthus fragrans Lour. Tree 1300– A paste of stem or bark is used in LC
3000 the treatment of boils, carbuncles,
whooping cough and retinitis
Syringa emodi Wall. ex Tree 2400
Royle
Asclepiadaceae
Marsdenia roylei Wight Climber 2100 The juice of the stem is used in the
treatment of gastric troubles and
peptic ulcers
Vincetoxicum auriculatum Herb 2100
(Royle ex Wight) Kuntze
Gentianaceae
Arnebia benthamii (Wall. Herb 3300 Flowering shoots are used in
ex G.Don) I.M.Johnst. preparation of syrup and jam which
cure fever, tongue and throat
infections, cardiac problems
Arnebia euchroma (Royle Herb 3650 Root extract is applied along with
ex Benth.) I.M.Johnst. hair oil will reduce hair fall
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 195
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Crawfurdia japonica var. Herb 2000–
luteo-viridis (C.B. Clarke) 3200
C.B. Clarke
Cynoglossum lanceolatum Herb 2300
Forssk.
Cynoglossum Herb 2300 Whole plant infusion is used to
microglochin Benth. cure cough and cold
Cynoglossum wallichii Herb 2800
G.Don
Cynoglossum zeylanicum Herb 2850 Decoction of leaves cures cold and
(Sw. ex Lehm.) Thunb. ex cough
Brand
Eritrichium canum Herb 2900
(Benth.) Kitam.
Gentiana albicalyx Herb 2350 Plant extract applied on cut wounds
Burkill.
Gentiana argentea Royle Herb 2400
ex D.Don.
Gentiana capitata Herb 3000
Buch.-Ham. ex D.Don.
Gentiana carinata Herb 3200
(D.Don ex G.Don)
Griseb.
Gentiana pedicellata Herb 700–
(D.Don) Griseb. 3800
Gentiana stipitata Edgew. Herb 2600 Plant juice used to cure fever
Gentiana tubiflora Herb 3500 Whole plant extract with curd is
(G.Don) Griseb. given to cure jaundice
Gentianella pedunculata Herb 2750 Plant extract is used to cure fever
(D.Don) H.Sm.
Hackelia uncinata (Royle Herb 3010
ex Benth.) C.E.C.Fisch.
Halenia elliptica D. Don. Herb 700–
4100
Lasiocaryum munroi Herb 3400–
(C.B.Clarke) I.M.Johnst. 4000
Lomatogonium Herb 3250 Plant cures liver and bile diseases
carinthiacum (Wulfen)
A.Braun
Swertia angustifolia Herb 2300 Decoction of whole plant cures
Buch.-Ham. ex D. Don fever
Swertia chirayita (Roxb.) Herb 1900 Plant treats liver disorders, malaria,
H.Karst. and diabetes
Swertia cordata (G.Don) Herb 2900 Plant shows antioxidant,
Wall. ex C.B.Clarke antibacterial and antidiabetic
activities
(continued)
196 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Swertia cuneata D. Don Herb 3200 Plant extract is given to cure fever
Swertia paniculata Wall. Herb 2800– Decoction of plant used in the
3300 treatment of malaria and other
fevers
Swertia purpurascens Herb 2500 Plant possess antibacterial and
(D.Don) C.B.Clarke anti-diabetic potential
Swertia speciosa D.Don Herb 2700 Plant is used in treating hepatitis,
cholecystitis, pneumonia,
osteomyelitis, dysentery, and
scabies
Tripterospermum volubile Climber 2300–
(D.Don) H.Hara 3100
Cuscutaceae
Cuscuta reflexa Roxb. Climber 2050 Plant is used in the treatment of LC
urinary disorders, muscle pain,
cough and also used as blood
purifier
Solanaceae
Nicandra physalodes (L.) Herb 2100 Plant is used in the treatment of
Gaertn. contagious disorders, toothache,
intestinal pain, impotence, fevers
Nicotiana rustica L. Herb 1000 Plant parts are used as a poultice
and a wash in the treatment of
rheumatic swelling, skin diseases
and scorpion stings
Solanum nigrum L. Herb 2000 Plant is used in ulcers, asthma, skin
diseases, whooping cough. The
juice from its roots is used against
asthma and
Solanum viarum Dunal Shrub 1000 Plant is used for the treatment of LC
cancer therapy, Addison’s disease,
rheumatism, chronic asthma, skin
disease, obesity, and leukaemia
Scrophulariaceae
Bonnaya ciliata (Colsm.) Herb 1500 Leaf juice is given after child birth LC
Spreng.
Euphrasia himalayica Herb 3000 Extract of whole plant is used to
Wettst. cure cuts and wounds
Euphrasia schlagintweitii Herb 3000 Plant cures eye diseases
Wettst.
Hemiphragma Herb 2500 Plant juice applied to cuts and
heterophyllum Wall. wounds. Fruits edible
Kashmiria himalaica Herb 3000
(Hook.f.) D.Y.Hong
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 197
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Lindenbergia grandiflora Herb 2050
Benth.
Lindenbergia indica (L.) Herb 500– Plant juice is given in chronic LC
Vatke 1500 bronchitis, skin eruptions
Majus japonicus (Thunb.) Herb
Kuntz.
Pedicularis bicornuta Herb 2800 Flowers are used in the treatment of
Klotzsch vaginal and seminal discharges
Pedicularis bifida Herb 2800
(Buch.-Ham. ex D.Don)
Pennell
Pedicularis gracilis Wall. Herb 3100
ex Benth.
Pedicularis hoffmeisteri Herb 2750 Root infusion is used to treat
Klotzsch flatulence and diarrhoea in cattle
Pedicularis megalantha Herb 2300–
D. Don 4200
Pedicularis pectinata var. Herb 3000 Plant used to alleviate stomach
rosea P.Agnihotri & T. pain, flatulence, intestinal problems
Husain and high blood pressure
Pedicularis porrecta Wall. Herb 3600–
ex Benth. 4500
Pedicularis punctata Herb 3400– Decoction of plant is given to expel
Decne. 3700 intestinal worms
Picrorhiza kurroa Royle Herb 3100 Root extract is used to cure severe EN
ex Benth. fever. Rhizomes are used as to treat
liver ailments
Scrophularia calycina Herb 3100
Benth.
Scrophularia himalensis Herb 2400
Royle ex Benth.
Torenia crustacea (L.) Herb 1500 Plant is used in treating bilious LC
Cham. & Schltdl. disorders, dysentery, amenorrhoea
and hepatitis. The powdered herb,
mixed with rice water, is drunk to
relieve diarrhoea, vomiting and
cholera
Verbascum thapsus L. Herb 2400 Leaf decoction is mixed with honey LC
to cure cough and other pulmonary
diseases
Veronica cana Wall. ex Herb 3300
Benth.
(continued)
198 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Veronica laxa Benth. Herb 2500 Plant is used in the treatment of
disorders of the nervous system,
respiratory tract, cardiovascular
system and metabolism
Orobanchaceae
Orobanche alba Stephan Herb 2700
ex Willd.
Gesneriaceae
Didymocarpus Herb 2000 Plant is used for respiratory
aromaticus D.Don problems and chronic asthma
Henckelia bifolia (D.Don) Herb 1000– Plant cures fever and skin allergy
A.Dietr. 2500
Henckelia pumila Herb 2200
(D.Don) A.Dietr.
Platystemma violoides Herb 2400
Wall.
Rhynchoglossum Herb 1800
obliquum Blume
Acanthaceae
Dicliptera bupleuroides Herb 2050
Nees
Dicliptera paniculata Herb 1000 The essential oil extracted from the
(Forssk.) I. Darbysh plant is used to cure tuberculosis.
The paste of the plant is used for
sprain and bone fracture
Justicia simplex D.Don Herb 2100 Plant used to treat fever, vomiting
Rostellularia procumbens Herb 2500 The leaf juice is used to treat
(L.) Nees ophthalmia, asthma, coughs,
rheumatism, back pain and
flatulence
Strobilanthes Herb 2600
atropurpurea Nees
Strobilanthes attenuata Herb 2400
(Wall. ex Nees) Jacq. ex
Nees
Strobilanthes Herb 2200 The root juice is used for the
penttemonoides (Nees) treatment of high fever. It is a good
T.Anderson fodder
Strobilanthes tomentosa Shrub 700–
(Nees) J.R.I Wood 2800
Strobilanthes wallichii Shrub 3000
Nees
Phrymaceae
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 199
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Phryma leptostachya L. Herb 4300 A tea made from the roots was
gargled to cure sore throats and was
drunk in the treatment of
rheumatism, fevers
Lamiaceae
Ajuga parviflora Benth. Herb 2400 Plant used in hypertension,
hepatitis, or inflammation of the
liver and jaundice
Callicarpa macrophylla Herb 2100 Plant cures tumour, polydipsia, LC
Vahl diarrhoea, dysentery, diabetes, fever
Clinopodium umbrosum Herb 2700 Leaf juice is applied to cuts and
(M. Bieb) C. Koch wounds
Clinopodium vulgare L. Herb 1400 Plant is used for healing wounds
and treating warts due to virus
infection
Coleus barbatus Herb 1600 The root is considered as analgesic,
(Andrews) Benth. ex G. ophthalmic, and febrifuge. It is very
Don useful in epilepsy
Elsholtzia ciliata (Thunb.) Herb 1500– An infusion is drunk, or vapour
Hyl. 3500 from the boiling decoction can be
inhaled, in the treatment of
common colds, fevers, headaches,
diarrhoea
Elsholtzia eriostachya Herb 3650 The seeds are chewed as a
(Benth.) Benth. treatment for coughs and colds
Elsholtzia fruticosa Herb 2300 Plant is used to cure bruises, head
(D. Don) Rehder ache, constipation, dysentery
Elsholtzia pilosa (Benth.) Herb 2500 Plant is used in the treatment of
Benth. common colds, fevers, headaches,
diarrhoea, oedema and oliguria
Elsholtzia strobilifera Herb 2350 Plant is used in the treatment of
(Benth.) Benth. colds, headaches, pharyngitis,
fever, diarrheoa, digestion disorder
Lamium album L. Herb 2400 Plant is used in the treatment of LC
fracture, hypertension, leucorrhoea,
paralysis, putrescence, trauma
Leucas lanata Benth. Herb 2200 The crushed leaves are used to treat
mild fevers, colds, rheumatism and
snake bites, roundworm infection
Micromeria biflora Herb 2100 A paste of the plant is used as a
(Buch.-Ham. ex D.Don) poultice to treat wounds. A paste of
Benth. the root is pressed between the jaws
to treat toothache
(continued)
200 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Nepeta eriostachya Herb 2300 Leaves extract is given for fever
Benth.
Nepeta govaniana (Wall. Herb 2500
ex Benth.) Benth.
Origanum vulgare L. Herb 2600 Leaves used against cold, for
digestive, and respiratory problems
Perilla frutescens (L.) Herb 600– An edible drying oil is obtained LC
Britton 2400 from seed. Seed is used in the
treatment of asthma, colds, nausea,
abdominal pain, food poisoning,
allergic reactions, bronchitis and
constipation
Phlomoides bracteosa Herb 2800 Powdered leaves are mixed in tea
(Royle ex Benth.) and used against cough and cold.
Kamelin & Makhm. Flowers are crushed and used
against toothache
Pogostemon benghalensis Herb 500– Leaves are utilized for relieving
(Burm.f.) Kuntze 1000 body pain, headache and fever
Prunella vulgaris L. Herb 2400 Leaves shows antibacterial, LC
antioxidant and anticancer activities
for the treatment of cardiovascular
diseases
Salvia hians Royle ex Herb 1800 Plant is a stimulant, used as a
Benth. remedy for dysentery. The pith of
tender stems is pickled
Salvia nubicola Wall. ex Herb 2400 Plant is used in the treatment of
Sweet. dysentery, boils, fall injuries,
hepatic problems and cancer
Stachys sericea Cav. Herb 2800 Plant is used for the treatment of
stress, skin inflammations,
gastrointestinal disorders, asthma
and genital tumours
Stachys splendens Wall. Herb 2400–
ex Benth. 3600
Thymus linearis Benth. Herb 2300– Seed extract is given for indigestion
3700 and stomach pain
Thymus serpyllum L. Shrub 1500– Plant is used in cough syrups, LC
2800 infusions, oil, tinctures and
infusions
Plantaginaceae
Plantago himalaica Pilg. Herb 2700 Leaves slightly bruised, to apply on
wounds. Oil from the seeds is
antimicrobial, for chronic diarrhoea
and shigellosis, a bacillary
dysentery
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 201
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Amaranthaceae
Achyranthes bidentata Herb 2400 Plant is used in the treatment of
Blume aching back and knees and asthenia
of the lower limbs
Amaranthus hybridus L. Herb 1300 Plant is used in the treatment of
intestinal bleeding, diarrhoea,
excessive menstruation
Cyathula capitata Moq. Herb 2500 A decoction of the aerial parts of
the plant are drunk as a treatment
for cough. An infusion of the whole
plant is taken as a remedy for fever
and dysentery
Cyathula tomentosa Herb 2300
(Schult.) Moq.
Chenopodiaceae
Chenopodium album L. Herb 2500 Plant is used in the treatment of
rheumatism, bug bites, sunstroke,
urinary problems, skin problems
Phytolaccaceae
Phytolacca acinosa Roxb. Herb 2000– The root is used in the treatment of
2700 urinary disorders, nephritis, oedema
and abdominal distension
Polygonaceae
Bistorta affinis (D.Don) Herb 2500– Paste of young shoot is given in
Greene 3800 empty stomach to cure dysentery
Bistorta amplexicaulis Herb 2500 Paste of rhizome is applied on sores
(D.Don) Greene and wounds
Bistorta macrophylla Herb 3000– Root paste is given to infants for
(D.Don) Soják 3700 stomach problems. Leaves paste is
applied on wounds
Bistorta vacciniifolia Herb 3300
(Wall. ex Meisn.) Greene
Bistorta vivipara (L.) Herb 4500 Plant is used in the treatment of
Delarbre abscesses, as a gargle to treat sore
throats and spongy gums and as a
lotion for ulcers
Fagopyrum cymosum Herb 1800– A decoction is used in the treatment
(Trevir.) Meisn. 2600 of traumatic injuries, lumbago,
menstrual irregularities, purulent
infections, snake and insect bites
Fagopyrum tataricum (L.) Herb 4400 Leaves are cooked. The sprouted
Gaertn. seeds are used in salads, or ground
into a powder and used as a cereal.
Edible oil is obtained from seeds
(continued)
202 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Fallopia pterocarpa Climber 2100
(Wall. ex Meisn.) Holub
Koenigia delicatula Herb 2600–
(Meisn.) H.Hara 3500
Koenigia mollis (D.Don) Herb 2050 Young shoots are cooked as a
T.M.Schust. & Reveal vegetable or pickled. The whole
plant is astringent
Koenigia nummulariifolia Herb 4000
(Meisn.) Mesicek &
Sojak
Koenigia polystachya Herb 3300
(Wall. ex Meisn.)
T.M.Schust. & Reveal
Koenigia rumicifolia Herb 3100– Young shoots and seeds are edible
(Royle ex Bab.) 3700
T.M.Schust. & Reveal
Oxyria digyna (L.) Hill Herb 3200 Decoction of young plant is used
for colic pain
Persicaria capitata Herb 2050 Plant treats urinary calculi and
(Buch.-Ham. ex D.Don) urinary tract infections
H.Gross
Persicaria chinensis (L.) Herb 1800– The juice of the plant is used in the
H.Gross 3300 treatment of eye diseases, eczema
of ears
Persicaria microcephala Herb 500– Young shoots are cooked as
var. sphaerocephala 3200 vegetable and condiment
(Wall. ex Meisn.) H.Hara
Persicaria nepalensis Herb 2700 Root extract is used in the treatment
(Meisn.) H.Gross of fevers. A paste of the root is used
as a poultice on fresh wounds
Persicaria sinuata (Royle Herb 3300
ex Bab.) H.Gross
Polygonum paniculatum Herb 3000 The infusion is used in the
L. treatment of stomach complaints
Polygonum recumbens Herb 1500–
Royle ex Bab. 4000
Rheum australe D.Don Herb 3400 The whole plant extract cures cuts
and wounds
Rheum moorcroftianum Herb 3250
Royle
Rheum webbianum Royle Herb 2500 Decoction of stem is given to cure
stomach pain
Rumex acetosa L. Herb 2900 Leaf extract applied on cut and
wounds
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 203
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Rumex nepalensis Spreng. Herb 3000 A paste of the root is applied to
swollen gums, pimples and
ringworm and applied externally to
relieve headache
Piperaceae
Peperomia tetraphylla Herb 2400 Plant treats kidney disorders
(Forst.) Hook. & Arn.
Lauraceae
Cinnamomum tamala Tree 1900 Plant used in the treatment of bad LC
(Buch.-Ham.) T.Nees & breath from mouth, black spots on
C.H.Eberm. face, tooth decay, swelling, cough
and also in complications of
tuberculosis
Litsea elongata (Nees) Tree 2300 The seeds are used as a source of LC
Hook.f. oil for industrial purposes. The
wood is used for construction,
furniture making
Machilus odoratissima Tree 1850 The wood is used in house LC
Nees construction and for furniture. Also
burned as firewood
Neolitsea cuipala Tree 2200 The oil from the fruit is used to
(D.Don) Kosterm. treat itchiness and other skin
complaints
Neolitsea umbrosa (Nees) Tree 2700 An oil obtained from the fruit is
Gamble used for burning
Thymelaeaceae
Daphne papyracea Wall. Shrub 2400
ex G.Don
Eleagnaceae
Elaeagnus umbellata Shrub 2400 Plant used in cardiac and LC
Thunb. respiratory diseases. Seed oil is
used for the treatment of pulmonary
infections, coughs
Hippophae rhamnoides Shrub 2000– A high-quality medicinal oil is LC
Serv. subsp. salicifolia 3700 extracted from the fruit and used in
(D.Don.) Serv. the treatment of cardiac disorders,
also to heal burns, eczema and
radiation injury
Hippophae salicifolia Tree 2700 The oil is made from the fruit is
D.Don used in cardiac disorders
Buxaceae
Buxus wallichiana Baill. Tree 2200 Leaves are useful in the treatment
of rheumatism and syphilis. A
decoction is used to combat
intestinal worms
(continued)
204 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Sarcococca hookeriana Shrub 2000 Plant used in the treatment of
Baill. stomach pain, rheumatism, swollen
sore throat and traumatic injury
Euphorbiaceae
Euphorbia maddenii Herb 2600
Boiss.
Euphorbia pilosa L. Herb 2200
Euphorbia prolifera Herb 500–
Buch.-Ham ex D. Don 2300
Euphorbia stracheyi Herb 2700
Boiss.
Flueggea virosa (Roxb. Shrub 2300 Plant is used in stomach problems, LC
ex Willd.) Royle dysentery, diarrhoea, eye problems
Leptopus cordifolius Shrub 1200–
Decne. 2100
Phyllanthus parvifolius Shrub 1700
Buch.-Ham. ex D.Don
Daphniphyllaceae
Celtis australis L. Tree 2100 The decoction can be used to LC
astringe the mucous membranes in
the treatment of diarrheoa,
dysentery, and peptic ulcers
Daphniphyllum himalense Tree 2200 A paste of the wood is applied as a
(Benth.) Mull.Arg. poultice to boils
Moraceae
Ficus auriculata Lour. Tree 1600 The plant helps to regulate blood LC
pressure and blood cholesterol
Ficus hederacea Roxb. Climber 2400 Wood is used as firewood
Ficus semicordata Tree 2100 Plant parts are used in leprosy, LC
Buch.-Ham. ex Sm. diarrheoa, headache, fever, earache,
ulcer, gastric problems and boils
Ficus subincisa Buch.- Tree 2100 Fruits edible LC
Ham. ex Sm.
Urticaceae
Boehmeria virgata subsp. Shrub 2050 A fibre is obtained from the bark is
macrophylla (Hornem.) of excellent quality, is used for
Friis & Wilmot-Dear making sacks, bags, rough clothes,
nets, rope
Boehmeria virgata var. Shrub 3000 An excellent quality of fibre is
macrostachya (Wight) obtained from the bark used for
Friis & Wilmot-Dear making sacks, bags, rough clothes,
nets, rope etc
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 205
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Boehmeria virgata var. Shrub 2100
scabrella (Dalzell &
A.Gibson) Friis &
Wilmot-Dear
Debregeasia longifolia Shrub 1500 Leaf juice is applied to scabies. LC
(Burm.f.) Wedd. Fruits are edible
Debregeasia saeneb Shrub 1800 The aerial parts of the plant are LC
(Forssk.) Hepper & dried and powdered, then mixed
J.R.I.Wood with mustard oil and then applied
topically as an antifungal for curing
skin rashes, dermatitis and eczema
Elatostema monandrum Herb 2800
(Buch.-Ham. ex D.Don)
H.Hara
Elatostema sessile Herb 1200– Poultice of leaves used for
J.R.Forst. & G.Forst. 2400 abdominal disorders, body pain.
Plant paste used on boils, pimples
and blisters. Tender leaves
consumed as vegetable. Shoots
boiled and eaten
Girardinia diversifolia Shrub 2200 The plant is a part of antidote to
(Link) Friis snake bites. Ash of the plant is
applied externally for the treatment
of ringworm and eczema
Gonostegia triandra Herb 2700 Plant is used to treat boils and
(Blume) Miq. abscesses, abdominal cramps in
females and leucorrhoea. Also used
to treat bone dislocations and
fractures
Laportea bulbifera Herb 3500 Young leaves cooked and used as
(Siebold & Zucc.) Wedd. spinach
Pilea racemosa (Royle) Herb 2300
Tuyama
Pilea scripta (Buch.- Herb 2200
Ham. ex D. Don) Wedd.
Pilea umbrosa Wedd. ex Herb 2500 Plant used in liver disorders,
Blume cancer, rheumatism and in skin
disorders
Pouzolzia zeylanica (L.) Herb 2200 Plant decoction is used to treat
Benn. cough, pulmonary tuberculosis,
sore throat, enteritis, dysentery
Urtica ardens Link Herb 2400– Plant is used for jaundice, sprain,
2700 fractures etc
Urtica dioica L. Shrub 2500 Plant is used in the control of LC
cardiovascular disorders especially
hypertension
(continued)
206 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Juglandaceae
Juglans regia L. Tree 2100 Plant helps to cure helminthiasis, LC
diarrheoa, sinusitis, stomach pain,
arthritis, asthma, eczema, scrofula,
skin disorders, and endocrine
diseases
Myricaceae
Myrica esculenta Tree 2300 Plant used to treat asthma, cough,
Buch.-Ham. ex D.Don chronic bronchitis, ulcers,
inflammation, anemia, fever,
diarrheoa, ear, nose, and throat
disorders
Betulaceae
Alnus nepalensis D. Don Tree 2100 LC
Betula alnoides Buch.- Tree 2200– Plant extracts is an antidote against LC
Ham. ex D.Don 2600 snakebites and is used to treat
dislocated bones
Betula utilis D.Don Tree 2800– Plant act as skin disinfectant, and LC
3700 cures diseases of the blood and ear,
convulsions, wound healing,
bronchitis, leprosy
Carpinus viminea Lindl. Tree 2100 Wood is used for making furniture, LC
ex Wall. articles of sports and weaving
shuttles
Fagaceae
Castanopsis tribuloides Tree 1700 The bark is a source of tannins and
(Sm.) A.DC. can be used as a dye. Seeds cooked
and eaten
Quercus floribunda Lindl. Tree 2000– The seeds are astringent and LC
ex A.Camus 2800 diuretic. They are used in the
treatment of diarrhoea, indigestion
and asthma
Quercus leucotrichophora Tree 1500– Plant is used to cure urinary
A.Camus 2400 infection, tooth ache and piles.
Leaves are used as an astringent
and in the treatment of diarrheoa.
Gum resin is used in stomach pain
Quercus semecarpifolia Tree 2400– Plant is used to treat acute LC
Sm. 3300 diarrheoa, dysentery and
haemorrhages. Also used as a
mouthwash to treat toothache or
gum problems and is applied
topically as a wash on cuts, burns,
various skin problems
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 207
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Salicaceae
Populus ciliata Wall. ex Tree 2500 A paste of the bark, mixed with the LC
Royle ash of cow dung, is used as a
poultice to treat muscular swellings
Salix calyculata Hook.f. Shrub 3300
ex Andersson
Salix denticulata Shrub 2500 Plant is used for malarial fever; the
Andersson leaf and seed as a diuretic, for
jaundice, cough, dropsy,
rheumatism, and urogenital
diseases
Salix flabellaris Shrub 3300 Decoction of bark is given to cure
Andersson fever
Salix fruticulosa Shrub 3000 Used as fodder
Andersson
Salix lindleyana Wall.ex Shrub 3200
Andersson
Orchidaceae
Calanthe plantaginea Terrestrial 2200 The dried and powdered rhizome is
Lindl. mixed with milk and taken as a
tonic and an aphrodisiac
Calanthe tricarinata Terrestrial 1500– Leaf paste is applied as a poultice
Lindl. 3300 to treat sores and eczema
Crepidium acuminatum Terrestrial 2300 Plant is used in the treatment of
(D.Don) Szlach. blood disorders, burning sensation
in the body, male sterility, fever,
dysentery, external and internal
haemorrhage and general weakness
Cypripedium elegans Terrestrial 2800 EN
Rchb.f.
Dactylorhiza hatagirea Terrestrial 3000 Tuber extract given to cure
(D.Don) Soó whooping cough and fever
Dendrobium amoenum Epiphytes 600–
Wall. ex Lindl. 2000
Dienia cylindrostachya Terrestrial 2500–
Lindl. 4000
Epipactis helleborine (L.) Terrestrial 2450
Crantz
Goodyera fusca (Lindl.) Terrestrial 2700
Hook.f.
Goodyera repens (L.) Terrestrial 2900 The infusion can be held in the LC
R.Br. mouth as a treatment for toothache.
The root and the leaves cure
bladder problems
(continued)
208 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Gymnadenia orchidis Terrestrial 2900
Lindl.
Herminium elisabethae Terrestrial 2700
(Duthie) Tang &
F.T.Wang
Liparis rostrata Rchb. f. Herb 1500– Tubers are used to treat stomach
3000 disorders and also used as a tonic
Malaxis muscifera Terrestrial 3000 Plant is used in sterility, VU
(Lindl.) Kuntze haemorrhages, dysentery, fever,
emaciation, burning sensation,
rheumatism and debility. Paste
applied externally for insect bites
Oberonia ensiformis Epiphytes 600–
(Sm.) Lindl. 1000
Oberonia micrantha Epiphytes 2500
Lindl.
Oberonia pachyrachis Epiphytes 2000
Rchb.f. ex Hook.f.
Oreorchis foliosa (Lindl.) Terrestrial 2500
Lindl.
Satyrium nepalense Terrestrial 2500 Plant is used as energetic tonic and
D. Don cure fevers
Spiranthes sinensis Terrestrial 2600 Tubers are used as tonic. Decoction LC
(Pers.) Ames of plants is used in intermittent
fever. Paste of stem and roots used
in sores
Zingiberaceae
Cautleya spicata (Sm.) Herb 2300 The juice of the rhizome is used in LC
Baker the treatment of stomach problems
Hedychium spicatum Sm. Herb 2200 Rhizome extract has antimicrobial
activity which used for making
various types of drugs including
anti cancerous drug and essential
oil
Hellenia speciosa Herb 1000 Rhizome is used to treat fever, LC
(J.Koenig) S.R.Dutta rashes, asthma, bronchitis, and
intestinal worms
Roscoea alpina Royle Herb 2000– Extract of whole plant is given for
4300 cough and cold. Dried powder of
leaves used in wounds and cuts of
cattle
Roscoea purpurea Sm. Herb 2600 Plant is used for the treatment of
diabetic, hypertension, diarrheoa,
fever, inflammation
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 209
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Nartheciaceae
Aletris pauciflora Herb 3200 The root is chewed as a treatment
(Klotzsch) Hand.-Mazz. for coughs and colds
Iridaceae
Iris domestica (L.) Herb 2500 The juice of the root is used to treat
Goldblatt & Mabb. liver complaints
Iris kemaonensis Wall. ex Herb 3200 Plant is used to treat ear pain and
D.Don weakening of the eyesight
Dioscoreaceae
Dioscorea bulbifera L. Climber 2050 Plant is used in the treatment of
piles, dysentery, syphilis, ulcers,
cough, leprosy, diabetes, asthma,
and cancer
Dioscorea deltoidea Wall. Climber 2400 Plant act as vermifuge and
ex Griseb. anthelmintic, to get rid of intestinal
worms
Amaryllidaceae
Allium stracheyi Baker Herb 3100 Dried flowers used to reduce blood
cholesterol levels, act as a tonic to
the digestive system and circulatory
system
Allium wallichii Kunth Herb 3300 The bulbs, boiled then fried in
ghee, are eaten in the treatment of
cholera and dysentery. The raw
bulb is chewed to treat coughs and
cold
Liliaceae
Asparagus curillus Shrub 2050
Buch.-Ham. ex Roxb.
Cardiocrinum giganteum Shrub 1500– The leaves are used as an external
(Wall.) Makino 2500 cooling application to alleviate the
pains of wounds and bruises. A
paste of the root is applied as a
poultice to treat dislocated bone
Clintonia udensis Trautv. Herb 3000 Young leaves and shoots are cooked
& C.A.Mey. as a vegetable
Disporum cantoniense Herb 1000– The juice of the roots is used in the
(Lour.) Merr. 3000 treatment of fevers. Tender leaves
and young shoots are cooked and
used as a vegetable
Fritillaria cirrhosa Herb 3200 The extract of root is used to cure
D.Don asthma
Lilium oxypetalum Herb 3500
(D.Don) Baker
(continued)
210 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Lloydia longiscapa Hook. Herb 3300
Maianthemum purpureum Herb 2800 Tender young leaves and shoots are
(Wall.) LaFrankie cooked as a vegetable
Ophiopogon intermedius Herb 2500 Plant is hacking dry coughs, dry
D. Don tongue, mouth, and constipation
Paris polyphylla Sm. Herb 1800– Roots are analgesic, antiphlogistic, VU
2800 antipyretic, antispasmodic,
antitussive, depurative, febrifuge
and narcotic. A decoction of the
roots is used in the treatment of
poisonous snake bites, boils and
ulcers, diphtheria
Polygonatum Herb 3200
geminiflorum Decne.
Polygonatum multiflorum Herb 2700 Plant is used in hair growth
(L.) All. promotion and hair blackening
Polygonatum Herb 2300 Plant is made into a salep, a
verticillatum (L.) All. strength giving food. Plant is
diuretic
Streptopus simplex D.Don Herb 2600
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Luzula campestris (L.) Herb 2600
DC.
Araceae
Acorus calamus L. Herb 2200 Rhizome paste is applied to treat LC
headache, skin diseases and
wounds. Powdered rhizomes are
used as insecticide
Arisaema intermedium Herb 2400– Plant is used to treat asthma,
Blume 3700 bronchitis, cold, cough, and
laryngitis
Arisaema jacquemontii Herb 2900 Roots may be cooked and eaten. LC
Blume The leaves are fermented before
being eaten
Arisaema propinquum Herb 2600 Plant exhibits anthelminthic
Schott property
Remusatia vivipara Herb 1200– Plant cures inflammation and
(Roxb.) Schott 2300 arthritis, and used as analgesic, for
disinfecting the genitourinary tract
and in the treatment of reddish
boils
Sauromatum diversifolium Herb 2400–
(Wall. ex Schott) 4200
Cusimano & Hett.
Eriocaulaceae
Eriocaulon nepalense Herb 2000 LC
J.D.Prescott ex Bong.
Cyperaceae
Bulbostylis capillaris (L.) Herb 2400 Plant is used to treat urinary tract LC
Kunth ex C.B. Clarke infections
Carex cardiolepis Nees Herb 2300–
3000
Carex cruciata Wahlenb. Herb 2300
Carex nivalis Boott Herb 3400
Carex norvegica Retz. Herb 1500– LC
2300
Carex nubigena D.Don. Herb 3000 Roots are cooked and eaten
Carex obscura Nees Herb 3200
Carex sanguinea L. var. Herb 1500
foliosa C.B. Clarke
Carex sanguinea Boott Herb 1300–
2500
Carex setosa Boott Herb 2700–
4800
(continued)
212 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Carex supina Willd. ex Herb 1300
Wahlenb.
Carex unciniiformis Herb 3600
Boeckeler
Cyperus cyperoides (L.) Herb 2200 The juice of the roots is used in the LC
Kuntz. treatment of coughs and fevers. The
ground-up rhizome is applied
topically to skin disorders
Erioscirpus comosus Herb 2200 The fibrous leaves are employed for
(Wall.) Palla making twine and cordage
Fimbristylis dichotoma Herb 2050 The culms are used to make an LC
(L.) Vahl inferior matting. Good soil binder
Poaceae
Agrostis pilosula Trin. Herb 2600 Fresh and dry aerial parts directly
palatable. Good fodder
Andropogon munroi Herb 2450 Good odder grass
C.B. Clarke
Apluda mutica L. Herb 2100 Leaf paste applied on wounds with
bleeding
Arthraxon lancifolius Herb 2100
(Trin.) Hochst.
Arundinella bengalensis Herb 2050
(Spreng.) Druce
Bothriochloa bladhii Herb 2100
(Retz.) S.T.Blake
Calamagrostis Herb 2600 LC
pseudophragmites (Haller
f.) Koeler
Calamagrostis Herb 3200
scabrescens Griseb.
Capillipedium assimile Herb 600–
(Steud.) A.Camus 2100
Cenchrus americanus (L.) Herb 2300
Morrone
Cenchrus orientalis Herb 2400 LC
(Rich.) Morrone
Chrysopogon gryllus (L.) Herb 2050 The plant can be used for thatching.
Trin An essential oil obtained from the
roots can be used as an adulterant
for rose oil
Cymbopogon distans Herb 2300 A lemon-scented essential oil is
(Nees ex Steud.) extracted from this species for
W. Watson medicinal and industrial purposes
Dactylis glomerata L. Herb 3800 Plant is used for tumours, kidney
and bladder ailments
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 213
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Digitaria ciliaris (Retz.) Herb 2100 Fodder grass
Koeler
Digitaria setigera Roth Herb 2500
Drepanostachyum Small tree 2100 Young shoots are cooked and used
falcatum (Nees) Keng f. as a vegetable. The leaves are used
as a roofing material
Eleusine indica (L.) Herb 2100 Plant is applied externally to open LC
Gaertn. wounds to stop bleeding. A poultice
of the leaves is applied to sprains
and back pains
Eragrostis nigra Nees ex Herb 2350
Steud.
Eulalia mollis (Griseb.) Herb 2200
Kuntze
Festuca rubra L. Herb 2900
Festuca valesiaca Herb 3000
Schleich. ex Gaudin
Glyceria tonglensis Herb 4100
C.B.Clarke
Himalayacalamus Shrub 2700 Young shoots are cooked and used
falconeri (Hook.f. ex as a vegetable
Munro) Keng f.
Lolium giganteum (L.) Herb 2600
Darbysh.
Melica scaberrima (Nees Herb 2700
ex Steud.) Hook.f.
Microstegium falconeri Herb 2400
(Hook.f.) Clayton
Microstegium nudum Herb 2050
(Trin.) A. Camus
Miscanthus nepalensis Herb 2400
(Trin.) Hack.
Muhlenbergia duthieana Herb 2000– Plant is used as fodder LC
Hack. 3600
Muhlenbergia huegelii Herb 900– Good fodder grass
Trin. 3000
Oplismenus compositus Herb 2050 Plant is antiseptic, astringent, LC
(L.) P. Beauv. demulcent for dropsy and dysentery
Oplismenus undulatifolius Herb 2100
(Ard.) P. Beauv.
Phleum alpinum L. Herb 3000 LC
Poa alpina L. Herb 3300
Poa annua L. Herb 3000 LC
(continued)
214 R. Manikandan et al.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Poa nepalensis (Wall. ex Herb 1900– Forage grass
Griseb.) Duthie 3000
Saccharum rufipilum Shrub 2300
Steud.
Stipa roylei (Nees) Mez. Herb 2640
Tenaxia cachemyriana Herb 3100
(Jaub. & Spach)
N.P.Barker & H.P.Linder
Thamnocalamus Shrub 2500
spathiflorus (Trin.) Munro
Tripogon filiformis Nees Herb 1000–
ex Steud. 4500
Trisetum clarkei (Hook. Herb 3000
f.) R.R.Stewart
Ephedraceae
Ephedra gerardiana Wall. Shrub 4000 Plant is used in the treatment for VU
ex Klotzsch & Garcke colds, coughs, bronchitis, asthma
and arthritis
Cupressaceae
Juniperus communis L. Shrub 3300 Plant is diuretic, anti-arthritis, LC
anti-diabetes, antiseptic as well as
used for the treatment of
gastrointestinal and autoimmune
disorders
Juniperus indica Bertol. Shrub 3400 Plant cures digestion problems LC
including upset stomach, intestinal
gas, heartburn, bloating and loss of
appetite
Taxaceae
Taxus wallichiana Zucc. Tree 2700 Plant is used to treat common cold, EN
cough, fever and throat pain
Pinaceae
Abies pindrow (Royle ex Tree 2700 Plant is a tonic for bronchitis, LC
D.Don) Royle haemoptysis, asthma, inflammatory
conditions, fever, hypoglycaemia
Abies spectabilis Tree 2800 Leaf juice is taken to treat asthma NT
(D.Don) Mirb. and bronchitis and is also given to
infants suffering from fever and
chest infection
Pinus roxburghii Sarg. Tree 2200 The wood is aromatic, deodorant, LC
haemostatic, stimulant,
anthelmintic, digestive, liver tonic,
diaphoretic, and diuretic
Selaginellaceae
Selaginella chrysocaulos Herb 2100
(Hook. & Grev.) Spring
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 215
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Loxogrammaceae
Loxogramme involuta Herb 2300
(D.Don) C.Presl
Loxogramme porcata Herb 2200
M.G.Price
Polypodiaceae
Drynaria mollis Bedd. Climber 2400
Drynaria propinqua Climber 2200
(Wall. ex Mett.) J.Sm. ex
Bedd.
Goniophlebium amoenum Herb 2300
(Wall. ex Mett.) Bedd.
Goniophlebium Herb 2400
fieldingianum (Kunze ex
Mett.) T.Moore
Goniophlebium lachnopus Herb 2500
(Wall. ex Hook.) Bedd.
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Lycopodiaceae
Huperzia verticillata Herb 1200– Plant is antioxidant, anti-
(L.f.) Trevis. 1900 inflammatory and
acetylcholinesterase inhibitory
activities in the neural system, to
prevent Alzheimer’s disease
Pteridaceae
Pteris aspericaulis Wall. Herb 2600 The paste of rhizome is applied in
ex J.Agardh treatment of cuts, boils, and
muscular swellings
Pteris cretica L. Herb 2100 Fronds are antibacterial, is made LC
into a paste and applied to wounds
Pteris wallichiana Herb 2500 Fresh leaves are crushed and
C.Agardh applied to stop bleeding and
healing of wounds
Hemiontidaceae
Adiantum capillus-veneris Herb 2050 Leaf extracts used in coughs and LC
L. throat infections
Coniogramme affinis Herb 2200 Plant shows antioxidant,
Hieron antimicrobial or anti-inflammatory
activities. It is used for the
treatment of various chronic and
infectious diseases
Trisetopsis aspera Herb 2500
(Munro ex Thwaites)
Röser & A.Wölk
Vittariaceae
Haplopteris flexuosa Herb 2200
(Fée) E.H. Crane
Aspleniaceae
Asplenium dalhousieae Herb 2100 Plant is antibacterial
Hook.
Asplenium ensiforme Herb 2200 Young fronds and underground
Wall. ex Hook. & Grev. stem are edible
Asplenium laciniatum Herb 2500 Plant is used in folk medicine
subsp. tenuicaule
(Hayata) Fraser-Jenk.
Asplenium yoshinagae Herb 2100–
subsp. indicum (Sledge) 2400
Fraser-Jenk.
Woodsiaceae
(continued)
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 217
Table 1 (continued)
Altitude
Binomial Life form (m) Ethno-medicinal uses Remarks
Woodsia elongata Hook. Shrub 2200
Aspleniaceae
Athyrium atkinsonii Bedd. Herb 2100
Athyrium fimbriatum Herb 2050
T.Moore
Athyrium schimperi Moug Herb 2100 The sporophylls possess anti-
ex Fee bacterial properties
Diplazium esculentum Shrub 2100 Plant is used for the treatment of LC
(Retz.) Sw. diabetes, smallpox, asthma,
diarrheoa, rheumatism, dysentery,
headache, fever, wounds, pain,
measles, and high blood pressure
Diplazium maximum Shrub 2100 Young fronds are edible
(D. Don) C.Chr.
Dryopteridaceae
Dryopteris cochleata Herb 2050 Plant is used to treat epilepsy,
(D. Don) C.Chr. leprosy, cuts, wounds, ulcers,
swelling
Dryopteris Herb 2400
redactopinnata Soumen
K.Basu & Panigrahi
Dryopteris wallichiana Herb 2500
(Spreng.) Hyl.
Polystichum squarrosum Herb 2200 The sporophyll extract of is used as
(D.Don) Fee an anti-bacterial agent. Fronds are
anti-rheumatic
Polystichum stimulans Herb 2100
(Kunze. ex Mett.) Bedd.
Davalliaceae
Davallodes pulchra Herb 2100
(D.Don) M.Kato &
Tsutsumi
Leucostegia truncata Herb 2100 Rhizome is used as antibacterial
(D.Don) Fraser-Jenk. and in the treatment of constipation
Blechnaceae
Woodwardia unigemmata Shrub 2300 The decoction of rhizome and
(Makino) Nakai fronds internally administrated in
dysentery. Dried rhizome is used as
purgative, fronds used in skin
diseases, and infertility
218 R. Manikandan et al.
3.2 EET Species
According to Walter and Gillett [61], over 60, 000 species have been evaluated for
conservation status as per the internationally accepted criteria, of which 34,000 are
classified as globally threatened with extinction. The Botanical Survey of India has
published Red Data Books [62] which contain information on 622 threatened plants.
Kandwal has developed an assessment system that prepares global Red List of
threatened species. IUCN Red List of Threatened Species, a comprehensive assess-
ment of the prevailing risk of extinction of thousands of plant and animal species.
The global IUCN Red List is updated on a regular basis and the latest version was
released in 2020 as version 2020–2 (IUCN Red List, 2020) [63]. According to the
criteria for critically endangered, endangered and vulnerable taxa are classified
mainly basing on reduction in population size, extent of occurrence and area of
occupancy.
Sunderdhunga glacier valley and its surroundings, the following Endemic,
Endangered and Threatened (EET) species have been reported, viz., Acer caesium,
Aconitum heterophyllum, Aconitum violaceum, Acorus calamus, Angelica glauca,
Berberis aristata, Cyananthus integer, Dactylorhiza hatagirea, Delphinium denu-
datum, Dioscorea deltoidea, Fritillaria roylei, Habenaria pectinata, Hedychium
spicatum, Holboellia latifolia, Jurinea dolomiaea, Malaxis muscifera, Meconopsis
aculeata, Morina longifolia, Nardostachys grandiflora, Parnassia nubicola,
Picrorhiza kurroa, Polygonatum verticillatum, Rheum webbianum, Rhododendron
anthopogon, Rhododendron campanulatum, Saussurea obvallata, Skimmia
anquetilia, Swertia alata, Thalictrum foliolosum, etc.
Among the threats to the indigenous flora of the valley, the most important
anthropogenic species, deforestation, cattle grazing, timber and fuel wood cutting,
soil erosion, construction schemes, plantation, plant collection, fire, tourist pressure
and the invasive species result in degradation of forests. In addition, environmental
factors, such as seismic activity or violent stress strokes and floods, chase landslides
or wash away top soil and then alter or destroy the vegetation. The prospective con-
servation of such unmatched scenic beauty of the floristic composition as well as
herbal wealth of the present study area is essential and also it is a home for a lot of
endemic plants and endangered animals which helps in maintaining genetic
diversity.
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 223
4 Conclusion
Several measures are underway for protection of the species reaching extinction and
endemic, endangered and threatened (EET) species. These measures include survey
and inventorization of plant resources in wild and to develop an accurate database
for their planning and monitoring purposes, the Protected Area Network (PAN) has
been created for in situ conservation which has been designated as species, habitats
and ecosystems oriented. Ex situ conservation of rare, endangered, threatened and
endemic species has been undertaken through botanic gardens, zoological parks,
gene banks, etc., and as per Biological Diversity Act, 2002, state-wise list of species
of plants which are on the verge of extinction along with guidelines to prohibit and
regulate their collection, rehabilitate and preserve these species have been proposed.
Acknowledgements The authors are thankful to the Director, Botanical Survey of India (BSI),
Kolkata, Head of Office (HoO), BSI, Northern Regional Centre, Dehradun, and HoO, BSI,
Southern Regional Centre, Coimbatore, for providing the facilities to carry out the said project.
References
19. Naithani BD (1984–1985) Flora of Chamoli, vol 1 & 2. Botanical Survey of India, Howrah
20. Kandwal M (2009) Grass Flora of Uttarakhand Unpublished Ph.D. thesis. HNB Garhwal
University, Srinagar (Garhwal), Uttarakhand
21. Duthie JF (1906) Catalogue of the plants of Kumaon and of the adjacent portion of Garhwal
and Tibet based on the collections made by Strachey and Winterbottom during the years
1846–49. Revised and supplemented by J.F. Duthie
22. Duthie JF (1906) The Orchids of North-Western Himalaya, Calcutta
23. Issar SK, Uniyal MR (1967) Orchids of Uttarakhand Himalayas. Indian Forest 67:713–716
24. Khullar SP (1997) An illustrated flora of Western Himalaya, vol 1 & 2. International book
distributors, Bishen Singh Mahendra Pal Singh Dehradun, p 1100
25. Osmaston AE (1927) A Forest Flora of Kumaon. Government Press, Allahabad, p 600
26. Pande P (1985) Flora of Almora District. Ph.D. Thesis, Kumaon University, Nainital, Vol. 1 & 2
27. Pande PC (1991) Observation on the vegetation of Almora district in Western Himalaya. J
Econ Taxon Bot 15(3):539–551
28. Pande PC (2001) Diversity of monocotyledonous flora of Almora and Bageshwar districts.
(Kumaon Himalaya). In: Pande PC, Samant SS (eds) Plant diversity of Himalaya. Gyanodaya
Prakashan, Nainital, pp 193–210
29. Pande PC (2010) Diversity of dicotyledonous flora of Almora and Bageshwar districts of
Kumaun Himalaya. In: Tewari LM, Pangtey YPS, Tewari G (eds) Biodiversity potentials of the
Himalaya. Gyanodaya Prakashan, Nainital, pp 197–228
30. Pangey YPS, Pande PC, Sharma SD (1984) New plant record for Garhwal and Kumaon
Himalaya from Almora. J Econ Taxon Bot 5:827–830
31. Rai ID, Singh G, Rawat GS (2017) Flora of Kedamath Wildlife Sanctuary, Western Himalaya:
a field guide, vol 393. Bishen Singh Mahendra Pal Singh, Dehradun
32. Randhwa MS (1970) The Kumaon Himalaya. Oxford & IBH Publishing Co., New Delhi
33. Rau MA (1975) High altitude flowering plants of West Himalaya, Howrah
34. Rau MA (1981) Western Himalayan Flora. In: Lall JS (ed) The Himalaya-aspects of changes.
Oxford University Press, New Delhi, pp 50–63
35. Rawat GS (1984) Studies on high altitude flowering plants of Kumaon Himalaya. Ph.D. Thesis,
Kumaun university, Nainital
36. Rawat JK, Sharma SK (1992) Conservation of biological diversity in the Uttaranchal. Ind
Forest 118:352–360
37. Singh GN, Chandra P, Vineet LM, Tewari, Singh Bisht MP (2019) Observation on the
Phyto-diversity of Sunderdhunga valley, Uttarakhand, Western Himalaya. Indian Forester
145(12):1166–1175
38. Uniyal BP, Balodi B, Nath B (1994) The grasses of Uttar Pradesh-a checklist. Bishen Singh
Mahendrapal Singh, Dehradun
39. Fosberg FR, Sachet MH (1965) Manual for Tropical Herbaria. Int. Bur. Pl. Tax. & Nom.,
Regnum Vegetabile (Vol. 39), Utrecht
40. Bridson D, Forman L (1998) The Herbarium handbook, third edn. (Repr. 1999). Royal Botanic
Gardens, Kew
41. Hajra PK, Balodi B (1995) Plant wealth of Nanda Devi biosphere reserve. Botanical Survey
of India, Kolkata
42. Uniyal BP, Sharma JR, Choudhery U, Singh DK (2007) Flowering plants of Uttarakhand- a
checklist. Bishen Singh Mahendrapal Singh, Dehradun
43. Lynch OJ (1992) Securing community based tenurial rights in the tropical forests of Asia-
an overview of current and prospective strategies. Issues in Development, World Resources
Institute, Washington
44. Dwivedi AP (1993) Forests- the ecological ramifications. Natraj Publishers, Dehra Dun
45. Leach G (1987) Household energy in South-east Asia. International Institute for Environment
and Development, London
46. Kothari A, Pandey P, Singh S, Variava D (1989) Management of National Parks and Sanctuaries
in India. Status report. Indian Institute of Public Administration, New Delhi
Checklist Flora of Sunderdhunga Valley, Western Himalaya, with Emphasis… 225
47. Champion HG, Seth SK (1968) A revised survey of the forest types of India. Govt. of India
Press, Delhi
48. Dhar U, Rawat RS, Samant SS (1996) Structural diversity and representativeness of forest veg-
etation in a protected area of Kumaun Himalaya, India: implication for conservation. Biodivers
Conserv 6:1045–1062
49. Rau MA (1974) Vegetation and phytogeography of Himalaya. In: Mani MS (ed) Ecology and
biogeography in India, The Hague, pp 247–280
50. Rawal RS, Bankoti NS, Pangtey YPS (1994) Broad community identification of high altitude
forest vegetation in Pindari region of Kumaun (Central Himalaya). Proc India Vat Acad B
60(6):553–556
51. Rikhari HC, Chandra R, Singh SP (1989) Patterns of species distribution and community along
a moisture gradient within and Oak Zone of Kumaun Himalaya. Proc Indian Nat Acad B
55:431–438
52. Singh JS, Singh SP (1987) Forest vegetation of Himalaya. Bot Rev 53(1):80–191
53. Singh JS, Singh SP (1989) Forest vegetation of the Himalaya. Bot Rev 53:80–192
54. Singh JS, Singh SP (1992) Forests of Himalaya. Gyanodaya Prakashan, Nainital
55. Tewañ JC, Singh SP (1985) Analysis of woody vegetation in a mixed oak forest of Kumaun
Himalaya. Proc Indian Natl Sci Acad BSI:332–347
56. Upreti N, Tewari JC, Singh SP (1985) The oak forests of the Kumaun Himalaya (India)1:
composition, diversity and regeneration. Mt Res Dev 5(2):163–174
57. CCRIMH (1973) Medicinal Flora of Certain Districts in Uttar Pradesh. Central Council for
Research in Indian Medicine and Homoeopathy (CCRIMH)-Monograph-4
58. Chandrasekar K, Rawat B (2011) Diversity, utilization and conservation of ethnomedici-
nal plants in Devikund – a high altitude, sacred wetland of Indian Himalaya. Med Plants
3(2):105–112. https://doi.org/10.5958/j.0975-4261.3.2.017
59. Rawat B, Sekhar KC, Gairola S (2013) Ethnobotanical plants of Sunderdhunga valley, Western
Himalaya, India - traditional use, current status and future prospect scenario. Indian Forester
139(1):61–68
60. Ved DK (2003) Conservation Assessment and Management Prioritization for the Medicinal
Plants of J & K, Himanchal and Uttaranchal. Proc. of a Regional Workshop held at Shimla
during May 22–25, 2003
61. Walter KS, Gillett HJ (1998) The 1997 IUCN red list of threatened plants. IUCN, Gland and
Cambridge, UK
62. Nayar MP, Sastry ARK (eds.) (1987, 1988, 1990) Red data book on Indian plants, vol 1–3.
Botanical Survey of India, Calcutta. (Repr. 2000)
63. IUCN (2020) IUCN Red List Categories and Criteria Version 3.1, Prepared by the IUCN
Survival Commission, IUCN, Gland, Switzerland. http://www.iucn-csg.org/index.php/red-
list-categories, 2001. Accessed 24 Dec 2020
Phytomedicines Used in Respiratory
Diseases by Traditional Healers
of Lakhimpur and Dhemaji Districts
of Assam, India
1 Introduction
study also aims at highlighting the ethnomedicinal uses of plant resources. Like
other deadly diseases, respiratory diseases are also a major cause of mortality
worldwide. In most of the developing countries like India, it causes a nationwide
burden. Pneumonia, asthma, lung cancer, tuberculosis, etc., are some common
respiratory diseases that have been causing concern since ancient times. In the pres-
ent-day scenario, due to so many reasons like air quality, smoking tobacco, etc., the
rate of these kinds of patients is on the rise. Children are the most susceptible targets
of respiratory illnesses. Pneumonia is reported as the leading killer of young chil-
dren all over the world, while asthma affects about 14% of children globally each
year and is the most common chronic disease among children and adults. Although
modern medicines have advanced the quality and lifestyle of humankind in many
countries in recent decades yet the cost of health care for respiratory diseases is an
increasing burden of all the leading and underdeveloped countries. For example, in
the United States, asthma alone costs about $18 billion annually [10]. So, there is an
urgent need for information about alternate complementary medicines like tradi-
tional ways of curing these diseases using surrounding plants. There are many effi-
cient medicines for these respiratory diseases in the present allopathic medical
system. But, in areas where the modern medical system is not easily available, peo-
ple happily use traditional systems to treat these diseases even today. The knowl-
edge of their ethnomedicine is very ancient, transferred orally for generations and
generated mainly through the trial-and-error method. It shares just a part of the vast
information regarding traditional uses of common plants by the people. So, the sur-
vey of medicinal plants becomes extremely important for researchers because it
gives information about a particular plant in a particular area. The northeastern part
of India is the huge storehouse of traditional knowledge as the life of numerous
tribal communities in this area is almost fully forest dependent. They collect almost
everything to survive from the floristic vegetation of their areas. Assam is one of the
eight states of Northeast India covered mostly by forest with innumerable forest
villages that are yet to avail the benefits of modern developmental facilities [11, 12].
With its boundless forests and hills, Assam is a home of abundant medicinal plants
with different individual tribal communities [5, 6]. Lakhimpur and Dhemaji dis-
tricts of upper Assam are also mostly inhabited by many tribal communities who
practise traditional plant-based medicines. However, a few such preliminary works
have been done in the area that is quite insufficient against the potentiality of the
region [13–17].
2 Methodology
2.1 Study Area
The survey was carried out in Lakhimpur (26°48′ N to 27°53′ N latitudes and 93°42′
E to 94°20′ E longitudes) and Dhemaji (27°05′27″ N to 27°57′16″ latitudes and
94°12′ to 95°41′ longitude) districts of Assam (Fig. 1) during 2018–2020.
Phytomedicines Used in Respiratory Diseases by Traditional Healers of Lakhimpur… 229
Fig. 1 Map of study sites, Lakhimpur and Dhemaji districts of Assam, India
The area is located on the northern bank of the river Brahmaputra and the north-
eastern corner of the Indian state of Assam, with an area of 2227 sq km for
Lakhimpur and 3237 sq km for Dhemaji districts. Both districts encounter hot and
humid summer, cold and dry winter with high precipitation during monsoon [18,
19]. Inhabitants of the area belong to different tribes and communities including
Mishing, Tiwa, Boro, Sonowal-Kachari, Deuri and Adibasi are living in both
the districts. While they are having their respective languages, the main communi-
cating language is Assamese. Earlier the entire area was covered under forest and
people were living mainly in the forest. Even today, apart from small township
areas, most of the people live in such villages and survive well in their tradi-
tional ways.
2.2 Data Collection
A random and regular household survey was carried out from August 2018 to March
2020 in different villages of the area selected randomly. A semi-structured question-
naire was prepared for the survey following Jain and Mudgal [20]. Traditional heal-
ers, elderly villagers of both sexes, were interviewed for their knowledge about
traditional medicinal plants used in treating Pneumonia and asthma.
230 P. Gogoi et al.
Plants are initially identified in their local language (Assamese) and voucher speci-
mens were collected for scientific identification. The specimens were then pro-
cessed into mounted herbarium sheets [21, 22]. Plants were identified referring to
literatures [23–25] and compared with the Herbarium of Arunachal University
(HAU-Herbarium). Updated names of plants were obtained from websites of the
world online organization on plant taxonomy [26, 27]. Specimens were deposited in
the HAU-Herbarium for future reference.
Quantitative Evaluation: For a better understanding of the collected information,
(i) relative frequency of citations (RFC) and (ii) fidelity level (FL) were calculated
following Umair et al. [28].
(i) Relative frequency of citations (RFC): It describes the local significance of each
species from a study area. RFC is calculated by the formula
RFC = FC/N (0 < RFC > 1), where FC is the number of informants citing
for a particular species and N is the total number of informants.
(ii) Fidelity level (FL): It is the percentage of informants mentioning a plant species
for a single disease. FL is calculated as
FL (%) = (Np/N) × 100, where Np is the number of informants that sug-
gested the use of a plant in a particular disease and N is the number of infor-
mants. A high FL value indicates the high use of a particular plant species in
treating a particular disease by the healers of that area.
Table 1 Plants recorded from Lakhimpur and Dhemaji districts of Assam for the treatment of
respiratory diseases such as pneumonia and asthma
Assamese FL
Plant names [Family]; field no. name Habit Status Disease (%) RFC
Acmella paniculata (Wall. ex DC.) Huhoni H Wi Pneumonia 1.25 0.025
R.K. Jansen [Asteraceae];
GPinki-119
Acorus calamus L. [Acoraceae]; Bosh H Wi Asthma 1.25 0.025
GPinki-062
Ageratum conyzoides L. Gundhua bon H Wi Pneumonia 2.5 0.062
[Asteraceae]; GPinki-031
Allium sativum L. Nohoru H Cu Pneumonia 1.25 0.025
[Amaryllidaceae]; GPinki-158
Alocasia macrorrhizos (L.) G.Don Nol kosu H Wi Pneumonia 1.25 0.012
[Araceae]; GPinki-169
Calotropis gigantea (L.) W.T. Aiton Palti S Wi Pneumonia 1.25 0.012
[Apocynaceae]; GPinki-200
Asplenium nidus L. [Aspleniaceae]; Biyonijupa H Wi Asthma 1.25 0.012
GPinki-075
Azadirachta indica A. Juss. Mohaneem T Wi Asthma 1.25 0.062
[Meliaceae]; GPinki-001
Capsicum annuum L. [Solanaceae]; Jolokia H Wi, Pneumonia 1.25 0.025
GPinki-022 Cu
Carica papaya L. [Caricaceae]; Omita Wi, Pneumonia 1.25 0.025
GPinki-035 Cu
Centella asiatica (L.) Urb. Bor maanimuni H Wi Pneumonia 3.75 0.162
[Apiaceae]; GPinki-011
Cinnamomum verum J. Presl Daalchini T Cu Pneumonia 1.25 0.037
[Lauraceae]; GPinki-112
Citrus x limon (L.) Osbeck Gulnemu S Cu Pneumonia 3.75 0.125
[Rutaceae]; GPinki-018 Asthma 1.25
Clerodendrum infortunatum L. Dhopat tita H, S Wi Pneumonia 1.25 0.05
[Lamiaceae]; GPinki-196
Croton caudatus Geiseler Hunghungi lota S Wi Pneumonia 2.5 0.025
[Euphorbiaceae]; GPinki-064
Curcuma caesia Roxb. Kola halodhi H Wi Asthma 1.25 0.012
[Zingiberaceae]; GPinki-089
Cynodon dactylon (L.) Pers. Dubori bon H Wi Pneumonia 1.25 0.037
[Poaceae]; GPinki-110
Cyperus rotundus L. [Poaceae]; Kera bon H Wi Pneumonia 2.5 0.025
GPinki-120
Deeringia amaranthoides (Lam.) Matuktuka C Wi Pneumonia 1.25 0.012
Merr. [Amaranthaceae]; GPinki-094
Drymaria cordata (L.) Willd. ex Ghuronia H Wi Pneumonia 2.5 0.075
Schult. [Caryophyllaceae]; laaijabori Asthma 1.25
GPinki-03
Elettaria cardamomum (L.) Maton Elichi H Cu Pneumonia 1.25 0.037
[Zingiberaceae]; GPinki-115
(continued)
232 P. Gogoi et al.
Table 1 (continued)
Assamese FL
Plant names [Family]; field no. name Habit Status Disease (%) RFC
Eleusine indica (L.) Gaertn Bobosa bon H Wi Pneumonia 1.25 0.025
[Poaceae]; GPinki-013
Ficus racemosa L. [Moraceae]; Dimoru T Wi Pneumonia 1.25 0.037
GPinki-087
Guilandina bonduc L. [Fabaceae]; Letaguti T Wi Pneumonia 5 0.187
GPinki-032 Asthma 2.5
Hellenia speciosa (J.Koenig) Jomlakhuti H Wi Pneumonia 1.25 0.087
S.R.Dutta [Costaceae]; GPinki-048
Heteropanax fragrans (Roxb.) Keseru T Wi, Pneumonia 1.25 0.012
Seem. [Araliaceae]; GPinki-093 Cu
Hydrocotyle sibthorpioides Lam. Horu H Wi Pneumonia 3.75 0.175
[Araliaceae]; GPinki-02 maanimuni
Hygrophila phlomoides Nees H Wi Pneumonia 1.25 0.012
[Acanthaceae]; GPinki-139
Kaempferia rotunda L. Bhumichampa H Wi Pneumonia 1.25 0.012
[Zingiberaceae]; GPinki-046
Leucas aspera (Willd.) Link Durun bon H Wi Asthma 1.25 0.1
[Lamiaceae]; GPinki-041
Macaranga denticulata (Blume) Murulia T Wi Pneumonia 3.75 0.075
Mull.Arg. [Euphorbiaceae];
GPinki-101
Mangifera indica L. Aam T Wi Pneumonia 1.25 0.025
[Anacardiaceae]; GPinki-067
Momordica dioica Roxb. Ex Willd. Bhatkerela C Wi, Pneumonia 1.25 0.012
[Cucurbitaceae]; GPinki-050 Cu
Myristica fragrans Houtt. Jaaifal T Cu Pneumonia 1.25 0.037
[Myristicaceae]; GPinki-113
Ocimum tenuiflorum L. Kola tulokhi H Wi Pneumonia 2.5 0.087
[Lamiaceae]; GPinki-107
Oldenlandia diffusa (Willd.) Roxb. Bonjaluk H Wi Pneumonia 3.75 0.087
[Rubiaceae]; GPinki-012
Oroxylum indicum (L.) Kurz Bhatghila T Wi Pneumonia 1.25 0.05
[Bignoniaceae]; GPinki-047
Oryza sativa L. [Poaceae]; Dhan H Cu Pneumonia 1.25 0.012
GPinki-200
Piper nigrum L. [Piperaceae]; Jaluk C Wi, Pneumonia 22.5 0.412
GPinki-004 Cu Asthma 5
Piper thomsonii (C.DC.) Hook.f. Aauni paan C Wi Pneumonia 1.25 0.025
[Piperaceae]; GPinki-014
Piper betle L. [Piperaceae]; Paan C Cu Pneumonia 3.75 0.075
GPinki-063 Asthma 1.25
Plumbago zeylanica L. Aagesita H Wi Asthma 1.25 0.012
[Plumbaginaceae]; GPinki-096
Potentilla indica (Andrews) Th. Gorokhia bon H Wi Asthma 1.25 0.012
Wolf [Rosaceae]; GPinki-201
(continued)
Phytomedicines Used in Respiratory Diseases by Traditional Healers of Lakhimpur… 233
Table 1 (continued)
Assamese FL
Plant names [Family]; field no. name Habit Status Disease (%) RFC
Psidium guajava L. Modhuri T Wi Pneumonia 5.00 0.15
[Myrtaceae]; GPinki-017
Ricinus communis L. Ara H Wi, Asthma 1.25 0.037
[Euphorbiaceae]; GPinki-034 Cu
Rubus alceifolius Poir. [Rosaceae]; Jetulipoka H, S Wi Pneumonia 2.5 0.087
GPinki-033
Scoparia dulcis L. [Plantaginaceae]; Meetha bon H Wi Pneumonia 1.25 0.05
GPinki-010
Sida rhombifolia (L.) [Malvaceae]; Hunboriyal H Wi Asthma 1.25 0.025
GPinki-143
Solanum violaceum Ortega Titabhekuri S Wi Pneumonia 1.25 0.012
[Solanaceae]; GPinki-043
Spondias pinnata (L.f.) Kurz Omora T Wi Pneumonia 1.25 0.012
[Anacardiaceae]; GPinki-066
Stephania rotunda Lour. Gumraaj C Wi Pneumonia 1.25
[Menispermaceae]; GPinki-102
Syzygium aromaticum (L.) Merr. & Lon S Cu Pneumonia 1.25 0.05
L.M.Perry [Myrtaceae]; GPinki-114
Tabernaemontana divaricata (L.) Kathana S Wi Asthma 2.5 0.037
R.Br. Ex Roem. & Schult.
[Apocynaceae]; GPinki-074
Tamarix dioica Roxb. ex Roth Jharu bon S Wi Asthma 1.25 0.012
[Asteraceae]; GPinki-168
Thunbergia coccinea Wall. Ex D. C Wi Pneumonia 1.25 0.012
Don [Acanthaceae]; GPinki-100
Typhonium trilobatum (L.) Schott Kosu H Wi Pneumonia 1.25 0.012
[Araceae]; GPinki-026
Urena lobata L. [Malvaceae]; Hunboriyal H Wi Pneumonia 2.5 0.025
GPinki-037
Vitex negundo L. [Lamiaceae]; Posotia S Wi Pneumonia 2.5 0.037
GPinki-044
Zanthoxylum nitidum (Roxb.) DC. Tezmui C Wi Pneumonia 1.25 0.062
[Rutaceae]; GPinki-059
Zehneria japonica (Thunb.) H.Y.Liu Belipoka C Wi Asthma 1.25 0.025
[Cucurbitaceae];
GPinki-053
Zingiber officinale Roscoe Aada H Cu Asthma 1.25 0.037
[Zingiberaceae]; GPinki-118
Ziziphus jujuba Mill. Bogori T, S Wi Pneumonia 1.25 0.012
[Rhamnaceae]; GPinki-065
Abbreviations used: Habit: C climber, H herb, S shrub, T tree. Status: Cu cultivated, Wi wild
Total number of informants/respondents = 80
234 P. Gogoi et al.
Calotropis R Equal amount of roots (200 g each) of both The paste is applied on
gigantea C. gigantea and C. gigantea and C. chest, once daily for 3
Capsicum R annuum are boiled in water and a decoction days in adults and once in
annuum is prepared. Grounded powder of two–four 48 h in children.
Piper nigrum F dry fruits of P. nigrum are added into the
decoction and made into a paste.
Scoparia dulcis R Roots of S. dulcis (150 g) and U. lobata About 1 ml of warm crude
Urena lobata R (50 g) are crushed together and juice are juice is orally
extracted by squeeze-press method, a little administered before
salt is added, warmed and the mixture is breakfast, once daily for 3
ready for treatment. days.
Solanum F Six fruits of S. violaceum, one seed of G. One teaspoon of the crude
violaceum bonduc, and root of P. thomsonii (of a juice is orally given on
Guilandina S plant) are grounded and pressed/squeezed empty stomach, once
bonduc and extract the juice. Dry grounded fruit daily consecutively for 3
Piper R powder (21 fruits) of P. nigrum is added to days.
thomsonii the juice mixture and warm.
Piper nigrum F
(continued)
Phytomedicines Used in Respiratory Diseases by Traditional Healers of Lakhimpur… 235
Table 2 (continued)
Part
Plants used Preparation Administration
Oroxylum B Bark of O. indicum (200 g) is grounded/ The crude mixture is
indicum pounded with a little water and juice is orally administered thrice
Piper nigrum F extracted. Dry powder of P. nigrum fruit daily before meal
(21 fruits) are added and the juice mixture consecutively for three
is ready for treatment. days.
Vitex negundo B About 200 g of bark of V. negundo, a About 3–4 tablespoons of
Rubus YS handful of young leafy twigs of R. crude mixture is orally
alceifolius alceifolius and P. guajava are grounded administered on an empty
Psidium YS and a crude mixture of juice is extracted by stomach daily,
guajava squeezing and pressing. A handful of P. continuously for three
nigrum powder is added to the crude juice days.
Piper nigrum F
mixture, and is administer.
Citrus x limon L Five leaves of C. limon, and five young One teaspoon of the juice
Ageratum YS shoots of A. conyzoides are grounded mixture is orally given on
conyzoides together and crude juice is extracted by empty stomach, once
Piper nigrum F squeezing & pressing and a handful of daily consecutively for 3
P. nigrum dry fruit powder is added to the days. (the preparation is
juice mixture. given only to adults)
Kaempferia Rh About 100 g of rhizome of K. rotunda is One teaspoon of the juice
rotunda grounded and juice is extracted by is given after meal, once
Piper nigrum F squeezing and pressing. Grounded powder daily for a week. (only for
of 1–2 dried fruits of P. nigrum is added to children below 5 years of
the juice. age. The Informants don’t
disclose the reason for not
applying to other age
groups)
Momordica T About 3–4 tubers (100 g approx.) are A teaspoon of the juice is
dioica pressed and juice is extracted and added a given on empty stomach,
little salt to it. daily for three days.
Croton YS A handful of young shoots of C. caudatus, Three tablets are given
caudatus P. guajava, Z. jujuba, S. pinnata, one seed thrice daily, before meal
Psidium YS of G. bonduc, 3 whole plants of consecutively for 3 days.
guajava C. rotundus, 3–4 leaves of M. indica and
Ziziphus jujube YS C. limon are grounded and a thick paste is
made. A handful of dry fruit powder of
Cyperus WP
P. nigrum is added to the paste and make
rotundus
tablets.
Guilandina S
bonduc
Mangifera L
indica
Citrus x limon L
Spondias YS
pinnata
Piper nigrum F
(continued)
236 P. Gogoi et al.
Table 2 (continued)
Part
Plants used Preparation Administration
Citrus x limon L A handful of all the ingredients except P. About (8–10) ml of crude
Piper nigrum F nigrum are mixed and juice is extracted by juice is given on empty
Clerodendrum YS grinding, squeezing and pressing. Dry fruit stomach, once daily
infortunatum powder of P. nigrum (21 fruits) is added to consecutively for 3 days.
the crude juice and ready to administer.
Croton YS
caudatus
Ageratum YS
conyzoides
Oryza sativa F
Zanthoxylum R A handful of root of Z. nitidum, a leaf of P. About 2 tablespoon of the
nitidum betle, young shoot of D. cordata crude juice extract is
Guilandina S (one shoot), a seed of G. bonduc are given on empty stomach,
bonduc grounded and by squeezing & pressing the once daily consecutively
Myristica F juice is extracted. Remaining ingredients for 3 days.
fragrans are added in a small quantities (0.5–1 fruit/
flower bud) to the crude juice mixture for
Syzygium FB
taste, warm a little.
aromaticum
Elettaria F
cardamomum
Piper betle L
Drymaria YS
cordata
Piper nigrum F
Cinnamomum B
verum
Urena lobata R Roots from one or two U. lobata plant(s), About (5–10) ml of the
Heteropanax B with handful of P. betle leaves and young juice is given on empty
fragrans shoot of V. negundo, 100 g (approx..) of stomach, once daily
Carica papaya B bark of H. fragrans and C. papoya, are consecutively for 3 days.
grounded and juice is extracted by
Piper betle L
squeezing and pressing. Dry fruit powder
Vitex negundo YS of P. nigrum (21 fruits) fruit with a little
Piper nigrum F salt is added to the crude mixture.
Deeringia R A handful of root of D. amaranthoides and About 1–2 tablespoon of
amaranthoides add dry fruit powder of P. nigrum (9 fruits) crude juice is given on
Piper nigrum F with a little salt and warm for a few empty stomach, once
minutes. daily till cured.
Ficus racemosa B About (50–100) g of bark of F. racemosa is About 1–2 tablespoon of
Piper nigrum F grounded and extracted in water by crude juice is given on
squeezing and pressing. Dry fruit powder empty stomach, once dail
of 21 fruits of P. nigrum is added to the for 3–4 days.
crude juice.
(continued)
Phytomedicines Used in Respiratory Diseases by Traditional Healers of Lakhimpur… 237
Table 2 (continued)
Part
Plants used Preparation Administration
Thunbergia L A handful of leaves of T. coccinea, tuber of About (3–5) ml of juice is
coccinea M. denticulate and small piece of S. given, once daily till
Macaranga L rotunda are grounded, and juice is cured.
denticulata extracted by squeezing-pressing. added dry
Stephania Tu fruit powder of P. nigrum (21 fruits) are
rotunda added to the juice mixture.
Piper nigrum F
Macaranga B The bark (200 g) of M. denticulate is About 2–3 teaspoon of
denticulate extracted by infusion for about 1 h, the crude juice is given on
Piper nigrum F extract filtrate is added with fruit powder of empty stomach, once
21 dry fruits of P. nigrum. daily consecutively for 3
days
Hygrophila YS Mix all ingredients (handful) except P. About 5–10 ml of the
phlomoides nigrum and extract the juice by crushing, crude juice is given on
Psidium YS squeezing and Pressing. Dry fruit powder empty stomach, once
guajava of P. nigrum (21 fruits) is added to the daily consecutively for 3
Rubus YS crude juice mixture. days.
alceifolius
Cyperus UN
rotundus
Ocimum YS
tenuiflorum
Guilandina S
bonduc
Piper nigrum F
Macaranga B A handful of all the ingredients except P. About 10 ml of the juice
denticulata nigrum are grounded and extracted the is given on empty
Psidium YS juice by squeezing and pressing. The juice stomach, once daily
guajava obtain is added with dry fruit powder of 21 consecutively for 3 days.
Cynodon WP fruits of P. nigrum and the mixture is ready
dactylon for use.
Hellenia St
speciosa
Piper betle L
Piper nigrum F
Acmella YS
paniculata
Oldenlandia WP
diffusa
Alocasia Tu A medium sized tuber (100–200) g of A tablespoon of the
macrorrhizos A. macrorrhizos is boiled in water with decoction is given after
Piper nigrum F 21–31 dry fruits of P. nigrum and the meal, twice daily
decoction is prepared. consecutively for 2–3
days.
Abbreviations used: Parts used: L leaf, F fruit, S seed, R root, B bark, T tuber, WP whole plant, YS
young shoot, Rh rhizome, St stem, FB flower bud, Fr frond; Extraction solvent: water; Method of
extraction: Fresh-Squeeze and Pressing
238 P. Gogoi et al.
Table 3 (continued)
Part
Plants used Preparation Administration
Acorus calamus Rh Dry rhizome powder of A. calamus Three tablets to be taken
is mixed with a little honey and thrice daily, till cured.
make tablets.
Tamarix dioica L A handful of T. dioica leaves are About 10 ml of the juice is
Piper nigrum F grounded using pestle and mortar given on empty stomach,
and crude juice is extracted, powder once daily consecutively for
of dry fruits (21 fruits) of P. nigrum 3 days.
is added to the juice, warmed and is
ready for use.
Potentilla indica WP Handful of the whole plant of About 10 ml of the juice is
P. indica is grounded using a pestle given on empty stomach,
and mortar and crude juice is once daily consecutively for
extracted by fresh-squeeze and press 3 days.
method and is ready for the ailment.
Abbreviations used: Parts used: L leaf, F fruit, S seed, R root, B bark, T tuber, WP whole plant, YS
young shoot, Rh rhizome, St stem, FB flower bud, Fr frond; Extraction solvent: water; Method of
extraction: Fresh-Squeeze and Pressing
practitioners also use the external mode of administration. Healers often recom-
mend a strict diet to the patients during the period of treatment like restrictions to
meat, fish, eggs, certain vegetables, etc.
Among the recorded plants, 48 species were found to be used in treating pneu-
monia (Table 2), while 18 species were used in asthma treatment (Table 3) with
some species such as P. nigrum, G. bonduc, C. limon, D. cordata, C. verum, E. car-
damomum, M. fragrans and S. aromaticum being used as common additives for
both the diseases during the preparation of crude medicines by various healers. A
similar ethnomedicinal survey related to respiratory disease was carried out in other
parts of India and reported similar plants such as O. tenuiflorum, L. aspera, Z. offi-
cinale, A. conyzoides, H. sibthorpioides, C. asiatica, O. diffusa, O. indicum, G. bon-
duc and P. Guajava [29–33] and for asthma L. aspera, P. nigrum [34] and A. calamus
[35]. However, in the present study, the use of Tamarix dioica and Potentilla indica
against asthma treatment and the use of Hygrophila phlomoides and Stephania
rotunda against pneumonia recorded from the region are new ethnobotanical
records.
4 Conclusion
The wild plants in Assam are in use in disease management in households and are
still popular. Often new ethnobotanical records also surface through similar field
studies, viz. Tamarix dioica and Potentilla indica (anti-asthma); Hygrophila phlo-
moides and Stephania rotunda (anti-pneumonia). Contrary to the anti-bacterial
240 P. Gogoi et al.
properties reported for Thunbergia coccinea [36] none of the informants cited the
species for pneumonia treatment from the region. Therefore, for understanding the
actual efficacy and for the scientific exploitation for the benefit of mankind phyto-
chemical analysis and biological validation is recommended for each of these
plants. Therefore, the present study will open up possibilities for further pharmaco-
logical research.
References
21. Bridson D, Forman L (1998) The herbarium handbook, 3rd edn. Royal Botanic Garden, Kew
22. Das AP (2021) Herbarium Techniques. In: Bhandari JB, Gurung C (eds) Instrumentation man-
ual. Narosa Publishing House, New Delhi, pp 78–94
23. Hooker JD (1872–1897) The Flora of British India, 7 Vols. L. Reeve& Co. Ltd., Ashford,
Kent. London
24. Kanjilal UN, Das A, Kanjilal PC, Purkaystha C, De RN, Bor NL (1934–1940) Flora of Assam,
vol I–V. Govt of Assam Press, Shillong
25. Barooah C, Ahmed I (2014) Plant diversity of Assam – a checklist of angiosperms & gymno-
sperms. ASTEC, Guwahati
26. www.plantsoftheworldonline.org
27. http://www.worldfloraonline.org
28. Umair M, Altaf M, Abbasi AM (2017) An ethnobotanical survey of indigenous medicinal
plants in Hafizabad district, Punjab-Pakistan. PLoS One 12(6):1–22
29. Dwivedi N, Dwivedi S, Dwivedi A (2015) Herbal remedies for respiratory diseases among the
natives of Madhya Pradesh, India. Am J Life Sci Res 3(2):158–162
30. Kala CP (2020) Medicinal plants used for the treatment of respiratory diseases in Uttarakhand
state of India. Stud Ethno Med 14(1–2):1–8
31. Bushi D, Bam K, Mahato R, Nimasow G, Nimasow OD, Tag H (2021) Ethnomedicinal plants
used by the indigenous tribal communities of Arunachal Pradesh, India: a review. Ethnobot
Res Appl 22(34):1–40
32. Yadav N, Ganie SA, Singh B, Chhillar AK, Yadav SS (2019) Phytochemical constituents and
ethnopharmacological properties of Ageratum conyzoides L. Phytoter Res 33(9):2163–2178
33. Acharyya BK, Sharma HK (2004) Folklore medicinal plants of Mahmora area, Sivsagar dis-
trict, Assam. IJTK 3(4):365–372
34. Singh RK, Lego YJ, Sureja AK, Srivastava RC, Hazarika BN (2021) People and plant: learn-
ing with Adi community on ethnomedicinal practices and conservation in Arunachal Pradesh,
India. IJTK 20(1):74–82
35. Rana CS, Tiwari JK, Dangwal LR, Gairola S (2013) Faith herbal healer knowledge document
of Nanda Devi biosphere reserve, Uttarakhand, India. IJTK 12(2):308–314
36. Sultana N, Das S (2019) Preliminary phytochemical screening and in vitro antimicrobial
activity of the leaf extract of Thunbergia coccinea (family-acanthaceae). Int J Curr Pharm
Res:111–114
Understanding Phytomedicinal
Gastronomic Culture of the Nagas
in Nagaland, India
1 Introduction
Nagaland obtained its statehood on December 1, 1963, and lies between 25°06’N
and 27°04’N latitude and 93°20′E and 95°15′E covering an area of 16,579 sq. km.
The state offers a scenic beauty of mountains and terrains inhabited by several eth-
nolinguistic tribal groups characterized by similar yet unique traditions and prac-
tices. Traditionally the Nagas lived a hunter–gatherer lifestyle but the transition
influenced mainly by religion, education, and modernization has led to a sedentary
phase with changes in beliefs and palatable flavor profiles. Its land supports a con-
siderable amount of crops like corn, pulses, fibers, potatoes, tobacco, oilseeds, sug-
arcane, millets, and rice along with abundant wild fruits and vegetables. Historically
Nagas were dubbed as inhabitants of thick forests frequenting the bazaar of
Sivasagar bartering salt, iron, and other items in exchange for their fine agricultural
produce [1]. Christianity is the established religion of the state with social structures
and practices burgeoned by their religious beliefs.
Traditional food gastronomy reflects the interactions between the environment
and local societies which results in the biocultural heritage of traditional food items
that help sustain the local traditional foods and implements food sovereignty [2, 3].
It plays the dual nature of prolonging the dynamics of present consumption and
tends to idealize the local feebly to promote economic sources for the community.
In this context, Barthes [4] suggested the dynamics related to traditional food gas-
tronomy as an idea to express the beliefs and opinions of the individual and social
ethical orientation equally. Nagas were once known as “warrior tribes” marked by
Lydia Yeptho and T. Ajungla contributed equally with all other contributors.
their bravery, but their bravery now manifests itself in the abominable cooking and
eating habits they create under the nose of their neighbors. The surprise and linger-
ing suspicion are palpable in the aroma of the fermented food products intriguing
celebratory chefs like Gordon Ramsay to broadcasting in shows like “gourmet goes
tribal.”
Cultural cuisine of local and traditional foods before modernization and industri-
alization provided a cultural identity among the societies [5]. The continuous inter-
action of cultures with the local ecosystems resulted in the traditional food systems
harboring indigenous knowledge over generations [6]. To recuperate the local gas-
tronomic condition, foodways pave way to an experienced progressive marginaliza-
tion that unites a community [7, 8] by analyzing the history and cultural roots of a
food product and also analyzing the value chains and productive structure of these
products [9].
Table 1 (continued)
Phytomedicinal benefits against
Main ingredients Method of preparation various diseases
Goosefoot Young leaves are cooked as curry Ulcer, blood purifier, liver
or boiled or fried with chilly problem
Taro, longevity spinach The outer skin of the yam is peeled Increases appetite in children
off and prepared as a vegetable
curry along with longevity spinach
leaves
Indian nightshade The fruits are prepared as a curry High blood pressure and fever
with fermented soybean and potato
or deep-fried in oil
Mint The leaves are prepared with chilly Stomach ache, indigestion
and tomato as chutney or cooked
with fermented soybean as a curry
Sweet basil The leaves are cooked with High blood pressure
fermented soybean and potato as a
curry or boiled or prepared as a
chutney
Roselle Fresh/dried roselle leaves are High blood pressure
prepared as a vegetable side dish
or simply boiled.
Edible mushrooms, Boiled with ginger and salt as a Tonic to prevent food poising and
Chinese Sumac powder, curry or a pinch of Chinese Sumac stomach problems. Powdered
ginger, powder is added while preparing Chinese sumac is used to treat
mushroom curry allergies or nausea and vomiting
Wax gourd Prepared as a vegetable curry or Diabetes
taken as a boiled side dish
Vegetable fern, tomato, The leaves are prepared along with Headache, fever
fermented soybean, potato, tomato, a spoonful of
chilly, and potato fermented soybean and chilly as a
vegetable curry
Bitter tomato The fruits are boiled or prepared as High blood pressure
a vegetable curry along with potato
Beefsteak plant, The seeds are crushed and cooked Constipation
fermented soybean, with a small amount of fermented
tomato, chilly, ginger, soybean with chilly, salt, ginger,
garlic, and taro stems garlic, and dried taro stems
Stalkless Elatostema The leaves are prepared as a Gastritis
vegetable side dish or used for
making rice porridge with small
amount of fermented soybean
Black turtle bean The seeds are boiled with salt and Mouth ulcer
cooked until soft
May chang, chilly The fruits of May chang are Vomiting and nausea
crushed with chilly and salt and
prepared as chutney
(continued)
Understanding Phytomedicinal Gastronomic Culture of the Nagas in Nagaland, India 247
Table 1 (continued)
Phytomedicinal benefits against
Main ingredients Method of preparation various diseases
Chinese Scallion, The bulbs are crushed and Fever and loss of appetite
tomato, and chilly prepared as a chutney with roasted
chilly and tomato
Ginger and fermented Ginger is crushed and the juice is Used as an appetizer and during
soybean squeezed, roasted dry red chilly fever
along with salt and fermented
soybean is added and prepared as a
chutney
Fragrant Caper Vine, The fruits are either sundried, High blood pressure and diabetes
chilly, tomato made into pickles, or prepared as a
chutney with chilly and tomato
Fish mint, tomato and The leaves are taken as raw or Heart problem, blood purifier,
chilly prepared as a chutney with tomato high blood pressure
and chilly
Sawtooth coriander The leaves are taken as raw, Increases appetite
prepared as chutney, or added in
curries as a garnish
Tree bean, potato, The legumes are prepared with Dysentery
tomato, and chilly potato as a curry or taken raw as a
salad or added in chutneys
Rice porridge with Rice is soaked, ground, and cooked Stomach ailments, diarrhoea,
longevity spinach with water; longevity spinach fever, gastritis
leaves leaves are added as a garnish.
Jobs tears The seeds are soaked, pounded, Indigestion
and cooked with water till it turns
soft and served as a porridge
Millet, rice It is cooked with rice and water or Cancer
pressurized until soft and eaten as
a porridge
Ginger and honey Ginger is pounded properly and Cold and cough
one teaspoon of the juice is boiled
with one teaspoon of honey
Toothache plant The flower heads are either chewed Toothache
or mashed and applied to the
affected area
Lime The leaves are crushed are inhaled Nausea, appetizer
and the juice is taken with water
and honey or sugar
Sweet potato The tubers are boiled or roasted Low blood pressure
Holy basil and hooker Crushed with salt and chilly Headache
leaves (optional) and the brine is taken
Himalayan mint, The flowers and leaves are added Headache, loss of appetite
potato, tomato, chilly, as a garnish in curry preparation
and garlic with potato, tomato and garlic. It is
also prepared with chilly and salt
as a chutney
248 L. Yeptho and T. Ajungla
Table 2 Various plants used as phytomedicinal food with their common name, botanical name,
family, and parts used
Common name Botanical name Family Parts used
Tomato Lycopersicon lycopersicum (L.) Solanaceae Fruit
H. Karst.
Chilly Capsicum frutescens L. Solanaceae Fruit
Ginger Zingiber officinale Roscoe Zingiberaceae Rhizome, leaves
Chinese Knotweed Polygonum chinense L. Polygonaceae Leaves
Garlic Allium sativum L. Amaryllidaceae Bulb, leaves
Potato Solanum tuberosum L. Solanaceae Tuber
Tulsi Ocimum tenuiflorum L. Lamiaceae Leaves
Longevity spinach Gynura procumbens (Lour.) Merr. Compositae Tender shoots,
leaves
Guava Psidium guajava L. Myrtaceae Leaves, fruit
Pomegranate Punica granatum L. Lythraceae Fruit
May chang Litsea cubeba (Lour.) Pers. Lauraceae Fruit
Hooker’s chive Allium hookeri Thwaites Amaryllidaceae Leaves, bulb
Passion fruit Passiflora edulis Sims Passifloraceae Leaves, fruit
Jobs tears Coix lacryma-jobi L. Poaceae Seeds
Millet Setaria italica (L.) P.Beauv. Poaceae Seeds
Chinese Scallion Allium chinense G.Don Amaryllidaceae Leaves, bulb
Indonesian lemon Zanthoxylum acanthopodium DC. Rutaceae Leaves, fruit
pepper
Fragrant Caper Vine Stixis suaveolens (Roxburgh) Pierre Capparaceae Fruit
Indian pennywort Centella asiatica (L.) Urb. Apiaceae Whole plant
Bitter gourd Momordica charantia L. Cucurbitaceae Fruit
Papaya Carica papaya L. Caricaceae Leaves, fruit
East Indian glory Clerodendrum glandulosum Lindl. Lamiaceae Leaves
bower
Redwool plantain Plantago erosa Wall. Plantaginaceae Whole plant
Goosefoot Chenopodium album L. Amaranthaceae Leaves
Fish mint Houttuynia cordata Thunb. Saururaceae Whole plant
Sawtooth coriander Eryngium foetidum L. Apiaceae Leaves
Taro Colocasia esculenta (L.) Schott Araceae Rhizome
Indian nightshade Solanum indicum L. Solanaceae Fruit
Tree bean Parkia timoriana (DC.) Merr. Leguminosae Fruit/pod
Mint Mentha spicata L. Lamiaceae Leaves
Holy basil Ocimum sanctum L. Lamiaceae Leaves
Roselle Hibiscus sabdariffa L. Malvaceae Flowers
Toothache plant Spilanthes acmella (L.) L. Compositae Young shoots
Stinging nettle Urtica ardens Link Urticaceae Young leaves
Cape periwinkle Catharanthus roseus (L.) G.Don Apocynaceae Leaves, flower
Lemon Citrus limon (L.) Osbeck Rutaceae Fruit
Chinese Sumac Rhus semialata Murray Anacardiaceae Fruit
Mango Mangifera indica L. Anacardiaceae Fruit
(continued)
Understanding Phytomedicinal Gastronomic Culture of the Nagas in Nagaland, India 249
Table 2 (continued)
Common name Botanical name Family Parts used
Sweet potato Ipomoea batatas (L.) Lam. Convolvulaceae Tubers
Wax gourd Benincasa hispida (Thunb.) Cogn. Cucurbitaceae Fruit
Vegetable fern Diplazium esculentum (Retz.) Sw. Athyriaceae Fronds
Tree tomato Solanum betaceum Cav. Solanaceae Fruit
Neem Azadirachta indica A.Juss. Meliaceae Leaves
Beefsteak plant Perilla frutescens var. crispa Lamiaceae Seeds, leaves
(Thunb.) H.Deane
Bitter tomato Solanum gilo Raddi Solanaceae Fruit
Stalkless Elatostema Elatostema sessile J.R.Forst. & Urticaceae Leaves
G.Forst.
Black turtle bean Phaseolus vulgaris L. Leguminosae Fruit/pod
Sweet basil Ocimum basilicum L. Lamiaceae Leaves
Pleasant Himalayan Elsholtzia blanda (Benth.) Benth Lamiaceae Flowers and
mint leaves
Fig. 1 (a-h) Various plants used as phytomedicinal foods. (a) Gynura procumbens, (b) Ocimum
tenuiflorum, (c) Coix lacryma-jobi, (d) Hibiscus sabdariffa, (e) Elsholtzia blanda, (f) Eryngium
foetidum, (g) Diplazium esculentum, (h) Solanum indicum
acceptable to the consumer by using raw materials that are locally available [15–
17]. The essential concept of Crouch’s [18] 3:12:120 ratio on group of people expo-
nentially creating and enforcing cultural innovation is headed by a posse of mothers
or women whose food choices and preferences form the basis of food culture, espe-
cially with regard to local and traditional food items that shaped the sustainability
of the diet. The essence of ensuing tradition from shaping culture at home to healing
the soul with food reflects a woman’s individual ability to nurture and nourish. The
sociocultural context heavily influences food choices [19], while the social identity
is also contextual as the individual’s daily engagement with the historical, social,
and political aspects considered to study identity [20]. Acculturation preserves the
original culture while acquiring the new practices, processes, and customs function-
ing within the main culture [21]. It explains the cultural and psychological change
or alteration as a result of merging between cultures [22] and encompasses the
domains of language, socioeconomic, and cultural values [23]. Ethnicity, migration
to urban areas, education, tribal community, political party representation, and
social support are some of the ways that influence the multicultural social policies
in Nagaland. The motivation for such social policies has been the desire to ensure
tribal harmony among the diverse tribes inhabiting the state.
Drawing from culture women creates an organic outgrowth of community living,
creating cultural ties between the various tribes, home, and individual level. Her
food choice provides a unique camaraderie between the food she creates and the
individual pressing toward healing. Her choices, her skills, each story unique to her,
and her kitchen form a gastronomic diversity toward healing through food. The
Naga community is woven by the thread of religious occasions, festivity, and visita-
tion by friends, family, and neighbors, and each occasion is celebrated by preparing
festive foods. Women are dexterous in the preparation of food combining creativity
and skills with community ties, social regulations, and ancestral knowledge bring-
ing about food with healing properties. The various dishes create a multiculturalism
facet among the Nagas represented by totemic or emblematic dishes signifying a
particular tribe. The food culture in Nagaland has emerged to be a product over
centuries interspersed by religion, education, and modernity striving to maintain its
cultural identity while also abandoning some like the use of alcoholic rice beverages
which were once an integral part of the diet culture. Their picking up of a sedentary
lifestyle now includes the utilization of resources and practice of pot culture and
garden cultivation in the kitchen backyard. Women create foodways connecting the
race, ethnicity, or gastronomic culture of a community and help expound the food
choices by paving itinerary food pathways of that community. Foods connoisseur
social identities and central to foodways are sharing the common goal of healing
while seeking to improve an individual’s well-being. Simultaneously the knowledge
is inherited yet also created to upkeep the culinary tradition. Their role is prolifer-
ated with social structures dictating her foodways while in return they act as culture
keepers passionately committed to intrahousehold dynamics powered by the tradi-
tional knowledge governing the food choices and practices required for a healthy diet.
Understanding Phytomedicinal Gastronomic Culture of the Nagas in Nagaland, India 251
4 Conclusion
Culture acts as a catalyst in the evolutionary process of humans and reveals the most
profound of meanings that humans carry – their rationale, their origins, and their
purpose. Culture links to build a person and the level of values and norms fixed by
culture is often what is highlighted in human rights discourse. To encounter heroism
in the traditions of patriotic and communal society, women act as cultural keepers
creating and enforcing a unique camaraderie through the memories of family and
historical stories, rendering an irresistible call to preserve the identity of traditional
food practices in a constantly evolving culture. The socioeconomic and consump-
tion patterns reveal the diet culture acting as phytomedicinal foods. Nutrition and
health benefit claims reflect their lifestyle rooted in rich ancestral knowledge to
strengthen the integrative force of healing. Thus, the traditional gastronomy of
Nagaland prolongs the dynamics of the ancient culture which is evident in the daily
course of preparing, serving, and gifting of food items that ties the society at an
individualistic level, uniting and defining a community interwoven by cultural cui-
sines and unique food flavor profiles.
Declarations Author’s contributions: Lydia Yeptho contributed to the documentation and con-
struction of the manuscript.
T. Ajungla supervised the work.
Acknowledgment: The authors would like to thank all the informants for sharing their valu-
able knowledge. They thank Mrs. Nihoβli, Miss Viseranuo Pesieye, Miss Rokonuo Kuotsuo, and
Miss Andy for their assistance during the interview.
Ethics Approval: Not Applicable.
Consent to Participate: All the authors have their consent to participate.
Consent for Publication: All the authors have their consent to publish their work.
Conflict of Interest: The authors declare no competing interests.
Availability of data and materials: Not applicable.
Funding: Not applicable.
References
1. Chophy GK (2021) The empire and the pearly gates. In: Mukherjee R (ed) Christianity and
politics in tribal India. Permanent Black, pp 68–69
2. Nabhan GP (2010) Ethnobiology for a diverse world: microbial ethnobiology and the loss of
distinctive food cultures. J Ethnobiol. https://doi.org/10.2993/0278-0771-30.2.181
3. Pieroni A, Pwaera L, Ghulam MS (2016) Gastronomic ethnobiology. In: Albuquerque UP,
Nóbrega Alves RR (eds) Introduction to ethnobiology. Springer International Publishing,
Switzerland, pp 53–62
4. Barthes R (1961) Pour une psycho-sociologie de l’alimentation contemporaine. Ann Econ Soc
Civiliz 16(5):977–986
5. Jordana J (2000) Traditional foods: challenges facing the European food industry. Food Res
Int 33(3):147–152
6. Kuhleinin HV (2009) Why are indigenous peoples’ food systems important and why do they
need documentation? In: Kuhnlein HV, Erasmus B, Spigelski D (eds) Indigenous peoples’
252 L. Yeptho and T. Ajungla
food systems: the many dimensions of culture, diversity and environment for nutrition and
health. Food and Agriculture Organization of the United Nations and Centre for Indigenous
Peoples’ Nutrition and Environment, Rome, pp 1–7
7. Fontefrancesco MF (2015) Il futuro dei Comuni minori. Etnografia di una trasformazione in
corso. Dada Riv Antropol Post-Glob 5:161–178
8. Fontefrancesco MF (2018) La luce alla fine del tunnel: sviluppo locale, offerta turistica e valori
locali. In: Corvo P, Fassino G (eds) Viaggi enogastronomici e sostenibilità. Franco Angeli,
Milano, pp 111–122
9. Fontefrancesco MF (2020) Traditional festive food and fragile aspirations of development in
Italy: the case of agnolotti pasta. J Ethnic Foods 7:2
10. Bentley A (2008) Introduction. Food and foodways, vol 16. Routledge, London, pp 111–116
11. Lyons D (2007) Integrating African foods: rural food and identity in Tigray, highland Ethiopia.
J Soc Archaeol 7(3):346–371
12. Germann Molz J (2007) Eating difference: the cosmopolitan mobilities of culinary tourism.
Space Cult 10(1):77–93
13. Kuhnlein HV, Receveur O (1996) Dietary change and traditional food systems of indigenous
peoples. Annu Rev Nutr 16:417–442
14. Tamang JP (2010a) Diversity of fermented foods. In: Tamang JP, Kailasapathy K (eds)
Fermented foods and beverages of the world. CRC Press/Taylor and Francis, Boca Raton,
pp 41–84
15. Tamang JP (2010b) Diversity of fermented beverages and alcoholic drinks. In: Tamang JP,
Kailasapathy K (eds) Fermented foods and beverages of the world. CRC Press/Taylor and
Francis, Boca Raton, pp 85–125
16. Tamang JP (2010d) Himalayan fermented foods: microbiology, nutrition and ethnic value.
CRC Press/Taylor and Francis Group, New York
17. Tibor D (2007) Yeasts in specific types of foods, chapter 7. In: Handbook of food spoilage
yeasts, 2nd edn. CRC Press, Boca Raton, pp 117–201
18. Crouch A (2008) Community. In: Crouch A (ed) Culture making, recovering our creative call-
ing. Inter Varsity Press Books, pp 239–246
19. Roudsari AH, Vedadhir A, Amiri P, Kalantari N, Omidvar N, Eini-Zinab H et al (2017) Psycho-
socio-cultural determinants of food choice: a qualitative study on adults in social and cultural
context of Iran. Iran J Psychiatry 12(4):241
20. Reddy G, Gleibs IH (2019) The endurance and contestations of colonial constructions of race
among Malaysians and Singaporeans. Front Psychol 10:792
21. Newman AJ, Sahak SZ (2012) Purchasing patterns of migrant groups: the impact of accultura-
tion on ethnocentric behaviors. J Appl Soc Psychol 42(7):1551–1575
22. Sam DL, Berry JW (2010) Acculturation: when individuals and groups of different cultural
backgrounds meet. Perspect Psychol Sci 5(4):472–481
23. Lopez-Class M, Castro FG, Ramirez AG (2011) Conceptions of acculturation: a review and
statement of critical issues. Soc Sci Med 72(9):1555–1562
Medicinal Plants in the Indian Traditional
Medicine and Current Practices
1 Introduction
Traditional medicine (TM) has been the world’s ancient form of treatment, used to
diagnose and reduce mental and physical ailments [1]. It is also known as comple-
mentary and alternative medicine, as well as ethnic medicine, in most of the coun-
tries. Natural products, such as microbes, marine organisms, plants and animals,
have been used in traditional medication since ancient period. Natural products have
a chemical diversity that has emerged over decades, resulting in a variety of biologi-
cal and therapeutic properties [2]. These substances have proven to be a valuable
resource for producing new chemical constituents as well as scaffolds. Natural
products are being used to fulfil the urgent need for effective pharmaceuticals with-
out any side effects and thus play a key role in the development of drugs for the
prevention/treatment of diseases/disorders [3].
In the study and development of current medications, TM seems to be too vital
to be overlooked. Traditional medicine has a wide range of application despite the
presence of scientifically validated drugs and therapies. A single plant or formula-
tion may have numerous phytochemical compounds, namely flavonoids, alkaloids,
polyphenol, terpenoids, alkaloids, etc. [4]. These compounds exert their activity
independently or synergistically to impart the desired therapeutic activity [5]. It is
important to note that many of the plant-based medications used in clinical practice
nowadays are all derived from folk medicine. Also, they have no/lesser side effects
compared to the currently available drugs in the market, and hence, people are pre-
ferring to incorporate herbal remedies in their lifestyle. Pharmaceutical drugs treat
symptoms produced by certain diseases as defined by scientific pathology, whereas
herbal therapy, on the other hand, focuses on assisting the body’s own healing pro-
cess. Medicinal plants are commonly used in traditional medicine and considered to
be beneficial; however, they may be toxic. If medicinal plant toxicity has been docu-
mented, it is due to geographical location, mistaken identity of the plants in which
they are supplied, and improper preparation or administration by untrained clini-
cians [6].
Various societies have evolved in documenting beneficial healing strategies of
TM to tackle a variety of health- and life-threatening disorders throughout history
[7]. Traditional medicine system includes Ayurveda, Unani, Siddha, Kampo, tradi-
tional Korean medicine (TKM), and Traditional Chinese Medicine (TCM). These
therapies make use of natural ingredients and have also been widely practised for
many years across the globe, prospering in an orderly regulated medical systems
[8]. These may have some drawbacks such as lack of proper formulations and lack
of clinical trials, but they are still an important reservoir of human knowledge [2].
Indian Traditional Medicine (ITM) is the medical system believed to have origi-
nated in India, which has an ancient heritage of traditional medicine. The materia
medica of India provides a great deal of information on the folklore practices and
traditional aspects of therapeutically important natural products [9]. ITM is based
on various systems including Ayurveda, Siddha, Unani, and Homeopathy. India is
considered as a hot spot of various medicinal plants and herbs. In this chapter, an
attempt has been made to understand the Indian traditional medicine (ITM) system,
the historical background, concepts, and differences amongst the various disciplines
in the ITM. Further, the integration of ITM into the current lifestyle, their usage,
their scope, and future prospects have also been discussed.
Table 1 (continued)
Name of the
medicine Origin Current role and scope in modern
system country Characteristics medicine and research
Homoeopathy Germany It is based on cures like the Many homoeopathy medicines are
and India principle available in the market and have
also been approved by the Indian
Government
Traditional Africa Traditional medicine doctors Most of the African population
African treat patients holistically. prefers traditional African
Medicine They generally seek to medicine
recombine the mental and Research is being carried out to
social equipoise of sufferers study the various pharmacological
according to social properties of African medicine
relationships and rules
Traditional Greece It has assimilated the The Traditional European
European principles of Ayurveda, medicines are being used by the
Medicine Unani, and Siddha European population. Research
needs to be carried out to
understand the mechanisms of
these medications
herbalism. Many plants are employed in traditional African medicine, and little is
known about their bioactive constituents [19]. Thousands of people in South Africa
rely on traditional medicine. It is estimated that 80 per cent of the population con-
sumes traditional medicines known as Muti. Muti is a term derived from the medici-
nal plant that is attributed to traditional herbal-, mineral-, and animal-based drugs.
The plant Muti is generally regarded as a long-term remedy for various treatments.
African traditional herbal remedies have a promising future that could be accom-
plished via collaboration, partnership, and transparency in practice, particularly
along with conventional healthcare personnel [20].
The Traditional European Medicine (TEM) is a combination of various systems
of medicine across the European continent with its origin beginning in ancient
Greece with Hippocrates. The various pharmacopoeias of different folk practitio-
ners contain numerous medicines made from leaves, roots, bark, and minerals found
in the nature. The TEM has assimilated concepts of Ayurveda, TCM, Unani, and
other traditional medicine systems [21]. However, it has only been partially incor-
porated into the modern medicine compared to other TMs.
Indian traditional medicine’s therapeutic knowledge has developed plenty of sig-
nificant traditional methods based on different concepts and theories. The ITM has
a unique distinction as it has six recognized medicine systems in this category.
These include Ayurveda, Siddha, Unani, Yoga, Naturopathy, and homeopathy [9].
Apart from these, there are tribal groups who practise their own ancient and folklore
medicine. For example, Maibaron is a medicinal system practised by the Meiteis,
and a tribal group from the Indian state of Manipur and a tribal group in Tamil Nadu
named Irular have developed their own folklore medicine [22]. The ITM is based on
the five elements theory and the three humoralisms theory [23]. The five elements
theory states that the all organisms in the world consist of the five elements which
are the earth (Prithvi), water (Jala), fire (Agni), air (Vayu), and ether (Akasha). The
three humoralisms theory suggests that the body is balanced by the three kinds of
doshas (tridoshas). These tridoshas are vata (gas), pitta (bile) and kapha (mucus).
Any imbalance in these doshas leads to diseases. All the medicine systems of ITM
have been formed on these above theories utilizing different herbs and medici-
nal plants.
India Traditional medicine system has a rich and diverse heritage of cultural tradi-
tions which use a variety of medicinal plants and herbs. The Western Ghats, the
north eastern region, and the Himalayan region are home to a rich source of medici-
nal plants [9]. These medicinal plants and herbs are the basis for more than 60% of
anticancer medications in the market or in therapeutic applications. Approximately
80% of antibacterial, immunosuppressive, cardiovascular, and anticancer medica-
tions are currently derived from plants. Over 70 per cent of the 177 anticancer medi-
cines that have been approved are based on natural compounds. Around 25% of the
258 R. Basu et al.
world’s prescription medications are derived from plants and there are nearly 121 of
them are in usage [24]. Ayurvedic medicine is thought to use between 1200 and
1800 distinct types of plants, whereas Indian folk therapists use about 7500 differ-
ent types of medicinal plants in various remedies. To regulate and standardize the
usage of traditional medicine, a new department, the Department of Indian System
of Medicine and Homoeopathy (ISM & H), was established in 1995. In 2003, ISM
& H was renamed AYUSH, which stands for Ayurveda, Yoga and Naturopathy,
Unani, Siddha, and Homoeopathy, as a separate Department within the Ministry of
Health and Family Welfare of India, India. The different sections of ITM are as
follows.
3.1 Ayurveda
Ayurveda is regarded as the world’s oldest known system of medicines. The Atreya
Inner Medicine School and the Dhanvantari Surgery School were the two branches
of Ayurvedic medicine that developed between 1500 BC and 1000 BC as one of the
four Upavedas [22]. The main characteristic of Ayurvedic medicine is holistic treat-
ment, which emphasizes the close relationship between both the brain and the body
[25]. The Ayurveda system utilizes processed natural drugs rather than derived or
synthetic compounds highlighting that maintaining the stability of all parts of the
human body is more important than eradicating external infectious agents. Ayurveda
employs the “Panchakarma” approach. Panchakarma treatment is used in a variety
of ways to promote body rejuvenation, detoxification, and longevity [26].
Panchakarma is made up of five karmas (activities) that are used to eliminate impu-
rities from the body. Virechan (purification by powder, paste, or hot water extract),
Rakta moksha (bloodletting), Vaman (force vomiting with medications), Nasya
(through nasal medication like smoking, oil, and decocting), and Basti (enema with
therapeutic oil) are the five karmas.
Ayurveda and biomedicine have different perspectives on how the human body
works. The tridoshas – pitta, vata, kapha – are the foundations of this system. It is
worth noting that vata, pitta, and kapha are responsible for both physiological as
well as psychological wellbeing [22]. The three doshas are in perfect equilibrium,
signifying homeostasis. Changes in one has an impact on the other two. The three
doshas have been used to categorize all clinical symptoms. Vata, for example, is
associated with dryness, lightness, weightlessness, coolness, roughness, minute-
ness, and mobility. Pitta refers to characteristics such as minor unctuousness, pen-
etrating, producing heat, lightness, foul odour, inducing movement, and liquidity and
kapha refers to cold, soft, stable, slow and heavy [27]. All symptoms, including
common cold, coughing, fevers, dysentery, oedema, and skin irritations, have
already been categorized as vata, pitta, or kapha. The diagnosis in Ayurveda is the
removal of toxins, preserve the body functions and remove the imbalance in the
body to prevent or reduce the manifestation of the disease [23].
Medicinal Plants in the Indian Traditional Medicine and Current Practices 259
3.2 Siddha
3.3 Unani
Unani medicinal approach was developed by the Greeks and later urbanized by
Arabs into a sophisticated medical discipline based on the Greek philosopher and
physician Hippocrates’ framework [34]. The four circumstances of living, which
include hot, sodden, cold, and dry, as well as the four humours of Hippocratic the-
ory, namely mucus, yellow bile, dark bile, and blood, underlie the unani medicine.
Unani treatment was combined with ITM during the reign of the Mughal Emperors
and is now used by a large portion of India’s population. Afal (capacities), Mizaj
(temperaments), Quo (resouurces), Arawh (spirits), Arkan (components), Aklath
(humours), and Anza (organs) are the seven levels according to Unani medicine
system [34, 35]. Unani medicinal approach is a holistic treatment that remarkably
treats a wide range of disease conditions, whether in single doses or multiple doses
and or in formulations. This approach shows effective treatment options for gastro-
intestinal, neurological, and heart diseases [36].
260 R. Basu et al.
3.4 Homeopathy
Homeopathy is a treatment practice that uses drug preparations whose actions cor-
relate to the disorder’s presentations (associated symptoms, clinical findings, and
pathological conditions) in the specific patient. Homeopathy was invented by the
German physician Hahnemann and is today used all around the world [37].
Homeopathy works on the principle of law of similar and/or “like cures like”. For
example, substance causing a disease symptom in larger quantity can be used to
treat the same symptoms with smaller doses [38]. Homeopathic medicines are pre-
scribed in very high dilutions, thereby making them non-toxic. Special pharmacies
prepare these medicines with a vigilant and careful process of agitation and dilution.
The treatment and medications in homeopathy are patient specific. The physician
understands the complete history of the patient which includes the physical and
mental fitness and the lifestyle. Depending on the assessment, smaller doses of
medicines are initially prescribed and when the results are visible, the infinitesimal
doses are given thereafter [39].
3.5 Naturopathy
Traditional medicine has a long history in India. India’s Materia Medica contains a
depth of information on traditional and folklore characteristics of therapeutically
useful natural ingredients [42, 43]. To meet the modern consumer’s needs, several
Medicinal Plants in the Indian Traditional Medicine and Current Practices 261
Table 2 List of different Indian traditional medicine system products available in the Indian market
Product Ingredient Properties Dosage
AYUSH HERBS PVT LTD
A-OXY (AMLA) Standardized Extract of Antioxidant, Natural Adult 1–2 caplets/
capsules Amla (Emblica Vitamin C Provider, caps thrice a day
officinalis) Rasayana with water, Child –
1 caplet/cap twice a
day or as directed
Rentone capsules Standardized extracts of Urinary tract infections, Adult- 1–3 caplets/
Didymocarpus kidney stones, Bladder tablets thrice a day
pedicellata, Saxifraga stones, Cystitis, Prostatitis, with water
ligulata, Rubia cardfolia, Urethritis, Nephritis, depending upon the
Achyranthes aspera, Prostrate hypertrophy patient’s condition.
Tribulus terrestris, Child (less than
Ocimum basilicum, 10 years of age) –
Crataeva religiosa, 1–2 caplets/tablets
Mimosa pudica, Dolichus twice a day with
biforus, Cyperus water or as directed
rotundus, Asphaltum
Arjuna capsules Standardized extract of Strengthen the weakened Adult 1–2 caplet/
Arjuna (Terminalia heart muscles tablet with water,
arjuna) thrice a day after
meal and there after
adjust the dose
according to
monitored blood
pressure or as
directed by your
health provider
Carditone Standardized extracts of Strengthens the weakened Adult- 1–2 caplets/
Rauwolfia serpentina, heart muscles, CHF, high tablet twice a day
Convolvulus pluricaulis, blood pressure, heart with water after
Terminalia arjuna, palpitation, irregular meal and there after
Tribulus terrestris, Rosa heartbeat, and angina adjusting the dose
centifolia, Boerhavia according to
diffusa, Inula racemose, monitored blood
Onosma bracteatum pressure or as
directed
(continued)
262 R. Basu et al.
Table 2 (continued)
Product Ingredient Properties Dosage
Hepo forte (bhui Standardized extract of Strengthens the liver, and Adult- 1–2 tablets/
amla) capsules – Bhumi amla (Phyllanthus helps in hepatitis B caplets with water,
jaundice support Amarus) 2–3 times a day
before meal,
Child- 1 tablet/
caplets twice a day
with water before
meal
Livertone Standardized extracts of All hepatic issues like 1–2 caplet thrice a
capsules – Silybum marianum, cirrhosis of liver, fatty day with water
hepatoprotective Boerhavia diffusa, liver, hepatitis, anorexia, before meals
Picrorhiza kurroa, other drug-induced
Eclipta alba, Phyllanthus diseases, etc. It works as a
amarus, Tephrosia liver cleanser and detoxifier
purpurea, Andrographis
paniculata, Swertia
chirata
Karela capsules Standardized extract of Useful in type 2 diabetes 1–3 caplets/tablets
diabetes support Karela (Momordica (depends upon the
charantia) blood sugar) thrice
a day with warm
water or as directed
Bio Gymnema – Standardized extracts of Useful in type 2 diabetes, 1–3 cap/tab
Sugar Gymnema sylvestre, weakness due to diabetes, (depends upon the
Metabolism Pterocarpus marsupium, complication like blood sugar) thrice
Support Ocimum basilicum, retinopathy, nephropathy, a day with water
Momordica charantia, obesity, etc. half an hour before
Azadirachta indica, meal or as directed
Salacia chinensis, Aegle
marmelos, Trigonella
foenum-graecum,
Syzygium cumini, Vinca
rosea
Immunity kit (for Flucomune caplets Ayush Pharmaceuticals Ashwagandha DS:
whole family Ashwagandha DS recently launched an 1 capsules: twice or
protection) capsules immunity kit to provide thrice a day
Haldi capsules adequate immunity for Haldi capsules: 1
individuals in the event of a capsule twice or
pandemic. This Immunity thrice a day
Kit boosts one’s immune Flucomune
system to protect the body Capsules- 1–2
from infection tablets twice/thrice
a day
Rakat tone – Standardized extracts of Loss of memory, 1-caplet twice a day
memory and Ginkgo biloba, Bacopa Alzheimer’s disease, poor with water or as
nervous system monnieri, Centella circulation, stroke, tinnitus, directed
support asiatica, Ashtavarga, asthma and all kinds of
Pueraria tuberosa, neuritis degenerative
Asparagus adscendens, neuromuscular changes
Withania somnifera
(continued)
Medicinal Plants in the Indian Traditional Medicine and Current Practices 263
Table 2 (continued)
Product Ingredient Properties Dosage
Kuff sooth –Standardized extracts of Allergic sore throat, Adult – 2 tea
cough and Ephedra vulgaris, laryngitis, allergic spoonful thrice a
respiratory Emblica officinalis, rhinorrhea, sinusitis, day. Child – 1
support Ocimum sanctum, smokers cough, bronchial teaspoonful thrice a
Terminalia bellirica, asthma, and chronic and day with warm
Adhatoda vasica, acute bronchitis water or as directed
Glycyrrhiza glabra, by your health
Pistacia integerrima, provide
Trikatu, Achyranthes
aspera, Zizyphus
vulgaris, Nilgiri oil,
Solanum xanthocarpum,
Viola odorata, Cichorium
intybus, Mentha spicata
THE HIMALAYA DRUG COMPANY
Shigru Leaf extract of Moringa Used in the treatment of 1 tablet twice daily
pterygosperma inflammatory disorders
Ashwagandha Root extract of Withania Enhances immunity by 1 tablet twice daily
somnifera strengthening the antibody
and strengthens the body
Arjuna Bark extract of Reduces coronary artery 1 tablet twice daily
Terminalia arjuna blood flow and protects the
heart muscle
Gokshura Fruit extract of Tribulus Helps in managing erectile 1–2 tablets twice
terrestris dysfunction daily
LIV 52 Extract of Chicory and Protects the liver from As per the advice of
Capparis spinosa infection and also restores the doctor
the functioning of the liver
Septilin Extract of Indian Has immunomodulatory, As per the advice of
Bdellium, Licorice and anti-inflammatory and the doctor
Tinospora Gulancha antioxidant properties
Diabecon GS Extract of Gymnema Has antidiabetic potential As per the advice of
sylvestre, and has the ability to the doctor
Pterocarpus marsupium, improve pancreatic and
Asphaltum punjabianum liver function
BAIDYANATH
Triphala Emblica officinalis, Aids in easy bowel 1–2 tablets daily
Guggulu Terminalia chebula, movements, digestion and
Terminalia bellerica, helps in relieving
Commiphora mukul constipation. Also helps in
alleviation of inflammation
Ayush kwath Ocimum sanctum, Immunity booster, and 1 tablet daily with
tablet Cinnamomum verum, helps in relieving cough water
Zingiber officinale, and respiratory distress
Piper nigrum
Mahasudarshan Tinospora Immunity booster, removal 3–6 teaspoonful
Kadha cordifolia, Picrorhiza of intestinal worms and with equal amount
kurroa, aids in proper digestion of water twice
Terminalia chebula
(continued)
264 R. Basu et al.
Table 2 (continued)
Product Ingredient Properties Dosage
Chandraprabha Tinospora Relieves intermittent 1–3 tablets daily
Bati cordifolia, urination, burning urination with milk
Curcuma longa and prevents UTI infection
PATANJALI AYURVED
Divya Jivanti, Water, Ksheera, Used in the treatment of 1 teaspoonful in
Jeevantyadi Ghrita, Prapaundarika, eye problems water
Ghrit kakoli, Kshirakakoli
Pippali, Lodhra,
Saindhava lavana,
Shatahva, Madhuka,
Draksha Sita
Daruharidra, Triphala
Divya Manjishtha Rubia cordifolia Used in wound healing, As advised by
Churna skin diseases, gouty physician
arthritis
Amvatari Ras Triphala, Guggul, Used in the treatment of As advised by
Chitrak mool, Erand tel, rheumatoid arthritis physician
Gandhak, Parad
As mentioned earlier, India is a rich and diverse source of different medicinal plants
and herbs. There are more than 7500 different medicinal plants which are used in
the various disciplines of the ITM. The formulation also changes in accordance to
the traditional medicine system. The Ayurveda medicines are available in tablet and
syrup forms, while the Siddha medicines are generally available in the form of paste
termed as “leha” and homeopathy medicines are in miniscule tablets. The therapeu-
tic properties depend on the composition and also the part from which the medicine
is prepared. There is never ending list of medicinal plants in India, some of which
are mentioned in Table 3 and a few of them have been discussed.
4.1.1
Camellia sinensis
Camellia sinensis L. is commonly known as Tea, which is the second most widely
consumed beverage on the globe. People have been consuming it frequently since
3000 BC. Tea is a beverage made from the leaves of Tea originated in China, then
expanded to India, Japan, Europe, and Russia [44]. It is a frequently utilized medici-
nal herb in trials throughout India, and it is prominent in several traditional medical
approaches such as Unani, Ayurveda, and Homeopathy. Tea is broadly classified
into the following groups: green, yellow, white, oolong, black, and dark tea. Clinical
studies have shown that tea has both therapeutic and preventive benefits for
Medicinal Plants in the Indian Traditional Medicine and Current Practices 265
Table 3 List of different medicinal plants used in the Indian traditional medicine system
Common
Plant name Part used Properties
Allium sativum Garlic Bulbs Lowering cholesterol, infantile
convulsion
Embelia ribes Vai vidang Seeds Piles, headache, skin disease, diabetes
(continued)
266 R. Basu et al.
Table 3 (continued)
Common
Plant name Part used Properties
Ocimum sanctum Tulsi Leaves Cough, cold, cancer, diabetes,
hepatoprotective, bronchial asthma
(continued)
Medicinal Plants in the Indian Traditional Medicine and Current Practices 267
Table 3 (continued)
Common
Plant name Part used Properties
Curcuma longa Haldi Rhizome Treats cancer, arthritis, antioxidant,
anti-septic
Rubia cordifolia Manjistha Stem Purifies blood, removes gall stone, treats
diabetic ulcer
(continued)
268 R. Basu et al.
Table 3 (continued)
Common
Plant name Part used Properties
Swertia chirayita Chirayita Whole Treats liver disorders, dyspepsia,
plant diarrhoea, and diabetes
Punica granatum Dadamchhal Rind of the Treats hyper acidity, chronic diarrhoea,
fruit dysentery, piles, sore throat, prostate
cancer
oxidative stress-related ailments such as cancer, type 2 diabetes, liver disease, and
cardiovascular disease [44]. It has pharmacological activities like antistroke activ-
ity, antiparkinson, anti-Alzheimer activity, anti-ageing activity, and anti-caries
activity. It is effective for weight loss and treating skin disorders. Tea contains
approximately 4000 bioactive constituents, such as polyphenols (catechins and fla-
vonoids), volatile oil, amino acids, polysaccharides, lipids, vitamin C, inorganic
components (manganese, fluorine, aluminium), and alkaloids (caffeine, theobro-
mine). Green tea seemed to have the largest antioxidant potential as well as a higher
concentration of polyphenols, particularly catechins. Furthermore, multiple investi-
gations have revealed that polyphenols and caffeine are the primary components in
tea accountable for diverse bioactivities and different organoleptic properties [45].
Caffeine is the primary component of Camellia sinensis and serves as a secondary
metabolite. When compared to other plant extracts, the methanolic extract of white
tea inhibited DPP-IV enzyme the most [46]. As a result, tea is an appropriate nutri-
tional source of natural antioxidants, particularly phenolic components, which have
the ability to be utilized in beverages or nutritional supplements.
Desmodium gangeticum
4.2
Desmodium gangeticum (L.) DC, also referred as Salparni or Sal Leaves Desmodium
or Salwan is a valuable medicinal herb that belongs to the legumes family (Fabaceae).
It is found throughout tropical as well as sub-tropical areas, and it is extremely fre-
quent in India’s lower Himalayan areas. It is also common in Tropical Africa, China,
Japan, Vietnam, Thailand, Indonesia, Cambodia, and northernmost part Africa.
D. gangeticum has been practised for centuries in traditional and folklore medicine
throughout India to cure a variety of diseases [47]. This medicinal plant is included
in many Ayurvedic formulations and is regarded as a “Master of Medicinal Plant” in
Ayurveda due to its extensive use in formulations [48]. D. gangeticum contains fla-
vonoids, polyphenols, alkaloids, steroids, terpenoids, phenylpropanoids, pterocar-
pans, coumarins, and volatile oil. Flavonoids, alkaloids, and pterocarpans are the
most important of these bioactive compounds. D. gangeticum exhibits significant
antioxidant, anti-inflammatory, anti-leishmanial, cardioprotective, anti-ulcer, anti-
diabetic, wound healing, hepatoprotective, and renal protective activities. An aque-
ous extract of D. gangeticum showed a significant DPP-IV inhibitory potential [46].
The toxicity of D. gangeticum extract was investigated in mice at various dosages
(50–2000 mg), with hyperactivity, grooming, convulsions, sedation, hypothermia,
and mortality being reported. No mortality was reported following oral administra-
tion of the maximum dose (2000 mg/kg) of extract. Furthermore, gangetin was
found to have no toxic effect up to 7 g/kg orally, indicating that it is rather safe and
no data of D. gangeticum toxicity have been reported in traditional medicine [49].
270 R. Basu et al.
Moringa oleifera
4.4
mental acuity, and bone density. It may help treat malnourishment, general fatigue,
breastfeeding women, menopausal symptoms, anxiety, and osteoarthritis [59].
M. oleifera contains a unique mixture of minerals, amino acids, and antioxidants
that are beneficial to both nourishment and ability to heal. The World Health
Organization (WHO) has recommended M. oleifera as a substitute to food sources
to address malnutrition. It has the highest amount of isothiocyanates and glucosino-
lates, and it also contains rhamnose. M. oleifera leaf, seed, and root extracts have
been shown in vitro and in vivo to have anticancer, hypoglycaemic, hepatoprotec-
tive, antibacterial, anti-inflammatory, antiviral, antifungal, and anti-sickling proper-
ties [60]. Leaf extracts have the highest antioxidant activity, and aqueous leaf
extracts have been shown to be safe in a variety of animal experiments. The reported
effects are thought to be caused by a variety of phenolic acids, polyphenols, as well
as glucosinolates, flavonoids, and possibly alkaloids. Up to a dose of 2000 mg/kg,
M. oleifera leaves (aqueous extract) were proven to be safe. 250 and 500 mg/kg
doses of the extract were administered intraperitoneally in fasting streptozotocin-
induced diabetic rats leading to remarkable reduction in blood glucose levels [61].
8 g of the powdered M. oleifera leaves per day for 40 days was administered to treat
type 2 diabetic individuals, and blood glucose during fasting and postprandial were
decreased by 28% and 26% in participants [61]. In terms of cost and efficacy, the
M. oleifera plant is the most cost-effective and dependable option for both healthy
nutrition, disease prevention, and cure.
Terminalia chebula
4.5
Tinospora cordifolia
4.6
Withania somnifera
4.7
Withania somnifera from the Solanaceae family, also called as Indian ginseng or
Ashwagandha, is widely available in India, Afghanistan, China, Africa, Yemen, and
Nepal. “Somnifera” in Withania somnifera in Latin is “sleep-inducer”, which justi-
fies its widespread use as a neuroprotective agent [69]. W. somnifera has been
widely used in Ayurveda, Siddha, and Unani medicine systems. Ashwagandha con-
tains over 35 phytochemicals that have been successfully isolated and
Medicinal Plants in the Indian Traditional Medicine and Current Practices 273
characterized. Alkaloids and steroidal lactones are the main chemical compounds
[70]. The alkaloids found in Ashwagandha include somniferine, somniferinine,
somnine, withanine, tropine, pseudo-withanine, pseudo-tropine, cuscohygrine,
3-a-gloyloxytropane, and anaferine, amongst which withanine is the most abundant
[71]. Phytopharmacological studies have proved that Ashwagandha has antioxidant,
anti-inflammatory, anticancer, antimalarial, antimicrobial, sedative, immunomodu-
latory, diuretic, and cardioprotective properties. Various researchers have examined
the use of Ashwagandha to treat Parkinson’s disease. A study by Bhatnagar et al.
2017 has proved that the ethanolic extract of Ashwagandha roots has been shown to
reverse Parkinson-like symptoms in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydrop
yridine) Parkinson induced Balb/c mice [72]. It has shown to restore behavioural
performance and significantly improve oxidative stress profile as a dose of 40 mg/
kg body weight. Ashwagandha has also been studied for its antidiabetic potential
wherein the phenolics present in the plant have been reported to reduce sugars levels
and Withaferin A is able to protect the islet cell damage through anti-inflammatory
property [73]. Withaferin A is also reported to have anticancer and antioxidant prop-
erty. Rasayana is a branch of Ayurveda which helps in promotion of body’s resis-
tance to diseases and increase the strength. W. somnifera is one of the prime rasayana
plants having a vast therapeutic property making it an excellent candidate to be used
in modern medications.
Bacopa monnieri
4.8
Ginkgo biloba
4.9
Ginkgo biloba, commonly known as ginkgo or the maidenhair tree, which is native
to Korea, China, and Japan, belongs to the family Ginkgoaceae. It is also found in
certain parts of India like Uttarakhand, Himachal Pradesh, West Bengal, Punjab,
and Meghalaya. It is used for weight loss effects along with antihypertensive, anti-
lipidaemic, and antidiabetic properties which may be effective in treating of meta-
bolic syndrome [77]. G. biloba nuts have long been used as food as well as medicine
throughout Asia. Because of their antibiotic properties, raw G. biloba nuts have
been used in Chinese traditional medicine to treat bacterial pathogens, asthma,
bronchitis, bladder, and kidney disorders [78]. Terpene lactones (bilobalide as well
as ginkgolides C, B, and A) and flavone glycosides (kaempferol, isorhamnetin, and
quercetin) are the main components of the plant [79]. Studies have shown that
G. biloba extract has successfully reduced body fat in overweight high-fat diet fed
rats. An eight-week study where the rats were administered 500 mg/kg body weight
of the extract as treatment has shown to minimize both body weight gain as well as
food intake compared to untreated rats [80]. The anti-inflammatory effect of the
plant extract may diminish the negative effects of high-fat diet consumption through
the reduction of TNF-retroperitoneal fat depot levels [77] Apart from this, G. biloba
is also known to have antioxidant, anticancer, and anti-inflammatory effects. The
polysaccharides of G. biloba have also studied for their medicinal properties such
as antioxidant, anticancer, anti-inflammatory, anti-depressant properties [81].
G. biloba is one of the oldest plants in the ITM which has captivated the human
beings for centuries through its promising application in traditional medicine.
Asparagus racemosus
4.10
According to WHO, 80% of world population rely and use traditional medicine.
Even though a large population of these are present in India, China, and Africa, the
developed countries have also started to explore the traditional medicine system
[22, 33]. Nature has been a constant source of medicinal herbs and plants. Earlier,
aspirin, digoxin, morphine, and quinine have been derived from plants [85]. The use
of phytotherapy is being used on a regular basis and the trend has again shifted to
the use of herbal remedies for the treatment of various diseases and disorders [86].
The ITM has also seen a rise in its usage in the last few decades [87]. The prac-
titioners of ITM have started addressing the core issues due to which the ITM indus-
try has started gaining importance [88]. More recently, isolating the compounds
from the medicinal plants and studying their pharmacological properties have
gained interest (Fig. 1, Table 4). The phytocompounds isolated have been reported
to work in targeted therapy. However, India had already started isolating compounds
from the medicinal plants in 1930s. Sen and Bose isolated two alkaloids from
Rauwolfia serpentina in 1931 [33]. In the same year, Siddiqui and Siddiqui isolated
five alkaloids, namely Ajmaline, Ajmalinine, Ajmalicine, Scrpentine, and
Serpentinine from the same plant [89]. In the years that followed, several other stud-
ies were also conducted by Indian researchers. During that time period, around 90%
of Indian doctors prescribed Rauwolfia serpentina as a daily antihypertensive, and
with manufacturers selling approximately 50 million tablets due to the work of
Rustom Jal Vakil [90].
Fig. 1 Structures of different phytoconstituents isolated from the medicinal plants of the Indian
traditional medicine system
276 R. Basu et al.
Fig. 1 (continued)
Medicinal Plants in the Indian Traditional Medicine and Current Practices 277
Fig. 1 (continued)
278 R. Basu et al.
Table 4 List of phytoconstituents isolated from some of the medicinal plants and their
pharmacological properties
Plant Plant part Phytoconstituent Pharmacological properties
Aegle Unripe Marmelosin Antidiarrhoea, aids digestion
marmelos fruits
Allium cepa Dried Alliin and Allicin Cardioprotective, antidiabetic, antibiotic
bulbs
Allium sativum Dried Alliin and Allicin Antibacterial, antigout, anti-inflammatory
bulbs
Aloe Gel Aloin and aloesin Anti-inflammatory, wound healing,
barbadensis Halophenols antidiabetic, immunomodulation
emodin Antidiabetic
Antidiabetic, anti-inflammatory, wound
healing, anticancer
Cassia Dried sennosides Purgative, aids digestion
angustifolia leaves
Centella Herb Asiaticoside and Antianxiety, coolant
asiatica Madecassoside
Curcuma Rhizome curcumin Anti-inflammatory, anticancer, antidiabetic,
longa antiseptic, wound healing,
immunomodulation
Echinacea Dried herb Alkylamide, Anti-inflammatory, immunostimulatory
purpurea echinacoside
Ferula Oleo gum Ferulic acid, Immunostimulatory, relieves flatulence,
asafoetida resin umbellic acid expectorant
Gingko biloba Leaves Ginkgolide, Antioxidant, stimulates platelets formation,
bilobalide treatment of migraine, and other
cerebrovascular diseases
Glycyrrhiza Root Glycyrrhizin and Antioxidant, anti-inflammatory, expectorant
glabra liquiritin
Hydrastis Dried root Berberine and Anti-inflammatory, antimicrobial,
canadensis hydrastine antihaemorrhagic, antioxidant, antidiabetic
Hypericum Dried Hypericin and Anti-inflammatory, anti-allergic,
perforatum aerial part hyperforin expectorant, antioxidant,
Panax ginseng Dried root Panaxosides, Immunostimulant, treatment of nervous
Ginsenosides system disorders, anticancer, antioxidant,
anti-inflammatory
Valeriana Dried root Valerate, Valerenic Tranquilizer, anti-inflammatory, relieves
officinalis acid pain and menstrual cramps, migraine
treatment
Zingiber Rhizome Gingerol, Immunostimulant, antioxidant, anticancer,
officinale Zingiberene antidiabetic, immunomodulation
Camellia Leaves Catechin Antioxidant, antidiabetic, anticancer
sinensis Epigallocatechin
Terminalia Fruits and Chebulinic acid Antioxidant, antidiabetic,
chebula leaves Β-sitosterol anti-inflammatory
Tinospora Leaves Magnoflorine Antidiabetic, immunomodulation,
cordifolia anticancer
Withania Leaves Somniferine Memory enhancer, neuroprotective
somnifera
Medicinal Plants in the Indian Traditional Medicine and Current Practices 279
The use of herbal medicines is booming and the market is growing tremendously as
sales of herbal medicines in India as over-the-counter products, ethical and conven-
tional formulations, and home treatments by ITM are approximately one billion
dollars, and export of plant extracts is about 80 million dollars [85]. Apart from this,
some Americans and Europeans come to India to enrol in courses and participate in
a comprehensive Indian medical treatment plan, which includes massage, food,
yoga, spiritual lectures, and meditation. This is turning into a unique marketing
feature for tourism in India. In addition, exports of medicinal plants and herbs have
been quite large in recent years. The main products of ITM from India are isabgol,
opium alkaloids, senna derivatives, Vinca rosa, Cinchona alkaloids, ipecac root
alkaloids, solasodine, diosgenin, menthol, gudmar grass, henna leaves, papian,
Rauwolfia guar gum, jasmine oil, sandalwood oil, etc. [96]. Indian pharmaceutical
companies and researchers with modern scientific knowledge and technology, and
ideas have begun rediscovering medicinal plants as a source of new drug candidates
based on traditional knowledge. Himalaya Drug Company, Dabur Limited,
Baidyanath, Patanjali Ayurved, Natural Remedies, etc., are some of the prominent
industries which have led to the introduction of ITM-based products in the Indian
market [94].
Modern technology and techniques have revolutionized the advancement of drug
research from the medicinal plants. Research using the latest equipment and meth-
ods have helped separate and develop plant components as new medicines present
in traditional herbal/medicinal plants formulations [22] (Fig. 2). Modern technol-
ogy monitors and maintains the quality of conventional formulations, using phyto-
chemicals as the basis for new drug discovery, determines pharmacokinetic profiles
and toxicity, and discovered mechanisms of existing herbal medicines [97]. It has
become an indispensable tool for finding new uses for prescribing, speeding up drug
discovery processes, synthetic and semi-synthetic processes for producing natural
ingredients, and more [98]. Traditional medicine is an important tool for discover-
ing new drugs as they are less expensive with minimal side effects and is readily
available locally. Quality control of marketed herbs/herbal formulations is critical
for obtaining maximum amount of phytochemicals/constituents for therapeutic effi-
cacy. As a result, herbal drug standards are essential. For comparing the quality of
herbal drugs, standard references such as the Indian Pharmacopoeia Reference
Substances, particularly botanical and phytochemical reference standards, are nec-
essary [99]. The Indian Pharmacopoeia Commission has now focused on standard-
izing the herbal products and promoting the usage of Indian traditional
medicine system.
Medicinal Plants in the Indian Traditional Medicine and Current Practices 281
Fig. 2 Prospect of drug discovery from the Indian traditional medicine system
7 Conclusion
References
1. Sewell RDE, Rafieian-Kopaei M (2014) The history and ups and downs of herbal medicines
usage. J Herbmed Pharmacol 3:1–3
2. Yuan H, Ma Q, Ye L, Piao G (2016) The traditional medicine and modern medicine from natu-
ral products. Molecules 21:1–28. https://doi.org/10.3390/molecules21050559
3. Li FS, Weng JK (2017) Demystifying traditional herbal medicine with modern approaches.
Nat Plants 3:1–13
4. Shi Y, Zhang C, Li X (2021) Traditional medicine in India. J Trad Chin Med Sci 8:S51–S55.
https://doi.org/10.1016/j.jtcms.2020.06.007
5. Gu R, Wang Y, Long B et al (2014) Prospecting for bioactive constituents from traditional
medicinal plants through ethnobotanical approaches. Biol Pharm Bull 37:1–15
6. Subramoniam A (2014) Present scenario, challenges and future perspectives in plant based
medicine development. Annals Phytomedicine 3:31
7. Walker AF (2006) Herbal medicine: the science of the art. Proc Nutr Soc 65:145. https://doi.
org/10.1079/pns2006487
8. Arentz S (2018) Developing intellectual capacity in naturopathy and herbal medicine practice.
Aust J Herb Naturop Med 30:6–7
9. Adhikari PP, Paul SB (2018) History of Indian traditional medicine: a medical inheritance.
Asian J Pharm Clin Res 11:421. https://doi.org/10.22159/ajpcr.2018.v11i1.21893
10. Zhang R, Zhu X, Bai H, Ning K (2019) Network pharmacology databases for traditional
Chinese medicine: review and assessment. Front Pharmacol 10:123. https://doi.org/10.3389/
FPHAR.2019.00123/BIBTEX
11. Park HL, Lee HS, Shin BC et al (2012) Traditional medicine in China, Korea, and Japan:
a brief introduction and comparison. Evid Based Complement Alternat Med 2012:429103.
https://doi.org/10.1155/2012/429103
12. Zhang J, Zhang Q, Liu G, Zhang N (2019) Therapeutic potentials and mechanisms of the
Chinese traditional medicine Danshensu. Eur J Pharmacol 864:172710
13. Kang YM, Komakech R, Karigar CS, Saqib A (2017) Traditional Indian medicine (TIM) and
traditional Korean medicine (TKM): aconstitutional-based concept and comparison. Integr
Med Res 6:105–113. https://doi.org/10.1016/J.IMR.2016.12.003
14. Kim JY, Pham DD (2009) Sasang constitutional medicine as a holistic tailored medicine. Evid
Based Complement Alternat Med 6:11–19. https://doi.org/10.1093/ecam/nep100
15. Yu F, Takahashi T, Moriya J et al (2006) Traditional Chinese medicine and kampo:
a review from the distant past for the future. J Int Med Res 34:231–239. https://doi.
org/10.1177/147323000603400301
16. Witt CM, Watanabe K, Matsuura K et al (2011) Traditional Japanese Kampo medicine: clinical
research between modernity and traditional medicine – the state of research and methodologi-
cal suggestions for the future. Evid Based Complement Alternat Med 2011:513842
17. Alrawi SN, Fetters MD (2012) Traditional arabic & islamic medicine: a conceptual model
for clinicians and researchers. Global J Health Sci 4:164–169. https://doi.org/10.5539/gjhs.
v4n3p164
18. Azaizeh H, Saad B, Cooper E, Said O (2010) Traditional Arabic and Islamic medicine, a
re-emerging health aid. Evid Based Complement Alternat Med 7:419–424. https://doi.
org/10.1093/ecam/nen039
19. Ezekwesili-Ofili JO, Okaka AN (2019) Herbal medicine in African traditional medicine. In:
Herbal Medicine
20. Mabona U, van Vuuren SF (2013) Southern African medicinal plants used to treat skin dis-
eases. S Afr J Bot 87:175–193
21. Micke O, Hübner J (2009) Traditional European medicine—after all, is Hildegard von Bingen
really right? Eur J Integr Med 1:226. https://doi.org/10.1016/j.eujim.2009.08.009
Medicinal Plants in the Indian Traditional Medicine and Current Practices 283
22. Pandey MM, Rastogi S, Rawat AKS (2013) Indian traditional ayurvedic system of medicine
and nutritional supplementation. Evid Based Complement Alternat Med 2013:376327. https://
doi.org/10.1155/2013/376327
23. Jaiswal A (2018) Traditional health care and traditional medicine in India. Archaeol
Anthropol:Open Access 2. https://doi.org/10.31031/aaoa.2018.02.000537
24. Sahoo N, Manchikanti P (2013) Herbal drug regulation and commercialization: an indian
industry perspective. J Altern Complement Med 19:957–963. https://doi.org/10.1089/
acm.2012.0275
25. Patwardhan B, Warude D, Pushpangadan P, Bhatt N (2005) Ayurveda and traditional Chinese
medicine: a comparative overview. Evid Based Complement Alternat Med 2:465. https://doi.
org/10.1093/ecam/neh140
26. Elahee S, Mao H, Shen X (2019) Traditional Indian medicine and traditional Chinese medicine:
a comparative overview. Chin Med Cult 2:105. https://doi.org/10.4103/cmac.cmac_29_19
27. Jaiswal YS, Williams LL (2017) A glimpse of Ayurveda – the forgotten history and principles
of Indian traditional medicine. J Tradit Complement Med 7:50–53
28. Sundarrajan S, Arumugam M (2017) Documentation of traditional Siddha medicines for skin
diseases from Katpadi taluk, Vellore District, Tamil Nadu. India Eur J Integr Med 9:52. https://
doi.org/10.1016/j.eujim.2016.08.163
29. Sieler R (2014) Patient agency revisited: “Healing the hidden” in South India. Med Anthropol
Q 28:323. https://doi.org/10.1111/maq.12067
30. Thas JJ (2008) Siddha medicine-background and principles and the application for skin dis-
eases. Clin Dermatol 26:62–78. https://doi.org/10.1016/j.clindermatol.2007.11.010
31. Patil SB, Patil MS, Chittam KP, Wagh RD (2014) A review on Ayurveda and Siddha: Indian
systems of medicine. Pharma Science Monitor 5:40–49
32. Vamshi Ram V, Premavathy D, Preetha S (2020) Assessing of knowledge of Siddha medi-
cine having antiviral property. Int J Pharm Res 12:575–583. https://doi.org/10.31838/
ijpr/2020.SP2.075
33. Sen S, Chakraborty R (2017) Revival, modernization and integration of Indian traditional
herbal medicine in clinical practice: importance, challenges and future. J Tradit Complement
Med 7:234–244
34. Islam A (2018) Origin and development of unani medicine: an analytical study. Intellectual.
Discourse 26:23–49
35. Rai N, Joshi SK, Sharma RK (2021) Regulatory requirements for quality control of unani
medicines. J AOAC Int 103:634–648. https://doi.org/10.5740/JAOACINT.19-0285
36. Itrat M (2020) Methods of health promotion and disease prevention in Unani medicine. J Edu
Health Prom 9:168
37. Manchanda R (2018) Experimentation in homoeopathy: history and prospects. Indian J Res
Homoeop 12:61. https://doi.org/10.4103/ijrh.ijrh_40_18
38. Cukaci C, Freissmuth M, Mann C et al (2020) Against all odds—the persistent popu-
larity of homeopathy. Wien Klin Wochenschr 132:232–242. https://doi.org/10.1007/
s00508-020-01624-x
39. Kaur H, Chalia DS, Manchanda RK (2019) Homeopathy in public health in India. Homeopathy
108:76–87. https://doi.org/10.1055/s-0038-1673710
40. Fleming SA, Gutknecht NC (2010) Naturopathy and the primary care practice. Prim Care Clin
Office Pract 37:119–136. https://doi.org/10.1016/j.pop.2009.09.002
41. Bradley R, Harnett J, Cooley K et al (2019) Naturopathy as a model of prevention-oriented,
patient-centered primary care: a disruptive innovation in health care. Medicina (Lithuania)
55:603. https://doi.org/10.3390/medicina55090603
42. Nugent-Head JA (2014) The first Materia Medica: the Shen Nong Ben Cao Jing. J Chin Med
43. Sharma H, Chandola HM, Singh G, Basisht G (2007) Utilization of Ayurveda in health care:
an approach for prevention, health promotion, and treatment of disease. Part 1 – Ayurveda, the
science of life. J Altern Complement Med 13:1011–1019
284 R. Basu et al.
44. Sharangi AB (2009) Medicinal and therapeutic potentialities of tea (Camellia sinensis L.) – a
review. Food Res Int 42:529–535
45. Saeed M, Naveed M, Arif M et al (2017) Green tea (Camellia sinensis) and L-theanine:
medicinal values and beneficial applications in humans—a comprehensive review. Biomed
Pharmacother 95:1260–1275
46. Sharma D, Kumar S, Kumar S, Kumar D (2019) DPP-IV inhibitors from natural sources: an
alternative approach for treatment and management of diabetes. Indian J Nat Prod Resour
10:227–237
47. Mehla J, Gupta P, Pahuja M et al (2020) Indian medicinal herbs and formulations for
Alzheimer’s disease, from traditional knowledge to scientific assessment. Brain Sci 10:964
48. Vedpal DSP, Dhamodaran P et al (2016) Pharmacognostical characterization, phytochemical
screening and finger print profile of the plant Desmodium gangeticum DC. Int J Pharmacogn
Phytochem Res 8:1271–1277
49. Bhattacharjee A, Shashidhara SC, Saha S (2013) Phytochemical and ethno-pharmacological
profile of Desmodium gangeticum (L.) DC.: a review. Int J Biomed Res 4:507. https://doi.
org/10.7439/ijbr.v4i10.355
50. Babu SN, Noor A (2020) Bioactive constituents of the genus Aloe and their potential thera-
peutic and pharmacological applications: a review. J Appl Pharm Sci 10:133. https://doi.
org/10.7324/JAPS.2020.101118
51. Babu SN, Govindarajan S, Vijayalakshmi MA, Noor A (2021) Role of Zonulin and GLP-1/
DPP-IV in alleviation of diabetes mellitus by peptide/polypeptide fraction of Aloe vera in
Streptozotocin- induced diabetic Wistar rats. J Ethnopharmacol 272:113949. https://doi.
org/10.1016/j.jep.2021.113949
52. Hamman JH (2008) Composition and applications of Aloe vera leaf gel. Molecules
13:1599–1616. https://doi.org/10.3390/molecules13081599
53. Boudreau MD, Beland FA (2006) An evaluation of the biological and toxicological properties
of Aloe barbadensis (Miller), Aloe vera. J Environ Sci Health Part C Environ Carcinogenesis
Ecotoxicol Rev 24:103–154
54. Aloe Chromones Improve Insulin Sensitivity.pdf
55. Yagi A, Hegazy S, Kabbash A, Wahab EA-E (2009) Possible hypoglycemic effect of Aloe vera
L. high molecular weight fractions on type 2 diabetic patients. Saudi Pharm J 17:209–215.
https://doi.org/10.1016/j.jsps.2009.08.007
56. Govindarajan S, Babu SN, Vijayalakshmi MA et al (2021) Aloe vera carbohydrates regulate
glucose metabolism through improved glycogen synthesis and downregulation of hepatic
gluconeogenesis in diabetic rats. J Ethnopharmacol 281:114556. https://doi.org/10.1016/J.
JEP.2021.114556
57. Mahmood KT, Mugal T, Haq IU (2010) Moringa oleifera: a natural gift-a review. J Pharm Sci
Res 2:775–781
58. Lakshmana Prabu S, Umamaheswari A, Puratchikody A (2019) Phytopharmacological poten-
tial of the natural gift Moringa oleifera lam and its therapeutic application: an overview. Asian
Pac J Trop Med 12:485–498
59. Sodhi G, Das S, Mazumder A, Rana S (2019) Ethnopharmacological review of drum stick
(Moringa oleifera): prevalently used as an alternative medicine. Res J Pharm, Biol Chem Sci
10:458–468
60. Sujatha BK, Patel P (2017) Moringa oleifera–nature’s gold. Imperial J Interdiscip Res
3:1175–1179
61. Stohs SJ, Hartman MJ (2015) Review of the safety and efficacy of Moringa oleifera. Phytother
Res 29:796–804
62. Rathinamoorthy R, Thilagavathi G (2014) Terminalia chebula – review on pharmacological
and biochemical studies. Int J PharmTech Res 6:97–116
63. Rao NK, Nammi S (2006) Antidiabetic and renoprotective effects of the chloroform extract
of Terminalia chebula Retz. Seeds in streptozotocin-induced diabetic rats. BMC Complement
Altern Med 6:17. https://doi.org/10.1186/1472-6882-6-17
Medicinal Plants in the Indian Traditional Medicine and Current Practices 285
64. Basha S, Reddy V, Sudha Rani Y et al (2017) A review on Terminalia chebula. Int J Pharmacol
Res 7:187–192. https://doi.org/10.7439/ijpr.v7i10.4431
65. Bag A, Bhattacharyya SK, Chattopadhyay RR (2013) The development of Terminalia chebula
Retz. (Combretaceae) in clinical research. Asian Pac J Trop Biomed 3:244–252. https://doi.
org/10.1016/S2221-1691(13)60059-3
66. Tiwari P, Nayak P, Prusty SK, Sahu PK (2018) Phytochemistry and pharmacology of Tinospora
cordifolia: a review. System Rev Pharm 9:70–78
67. Chi S, She G, Han D et al (2016) Genus Tinospora: ethnopharmacology, phytochemistry, and
pharmacology. Evid Based Complement Alternat Med 2016:9232593
68. Prince PSM, Menon VP, Gunasekaran G (1998) Hypolipidaemic action of Tinospora cor-
difolia roots in alloxan diabetic rats. J Ethnopharmacol 64:53–57. https://doi.org/10.1016/
S0378-8741(98)00106-8
69. Zahiruddin S, Basist P, Parveen A et al (2020) Ashwagandha in brain disorders: a review of
recent developments. J Ethnopharmacol 257:112876
70. Namdeo AG, Ingawale DK (2021) Ashwagandha: advances in plant biotechnological
approaches for propagation and production of bioactive compounds. J Ethnopharmacol
271:113709
71. Dhawan M, Parmar M, Sharun K et al (2021) Medicinal and therapeutic potential of withano-
lides from Withania somnifera against COVID-19. J Appl Pharm Sci 11:006–013. https://doi.
org/10.7324/JAPS.2021.110402
72. Bhatnagar M, Goel I, Roy T et al (2017) Complete Comparison Display (CCD) evaluation
of ethanol extracts of Centella asiatica and Withania somnifera shows that they can non-
synergistically ameliorate biochemical and behavioural damages in MPTP induced Parkinson’s
model of mice. PLoS One 12:e0177254. https://doi.org/10.1371/JOURNAL.PONE.0177254
73. Paul S, Chakraborty S, Anand U et al (2021) Withania somnifera (L.) Dunal (Ashwagandha): a
comprehensive review on ethnopharmacology, pharmacotherapeutics, biomedicinal and toxi-
cological aspects. Biomed Pharmacother 143:112175
74. Mathur D, Goyal K, Koul V, Anand A (2016) The molecular links of re-emerging therapy: a
review of evidence of Brahmi (Bacopa monniera). Front Pharmacol 7:44
75. Srivastava P, Raut HN, Puntambekar HM, Desai AC (2012) Stability studies of crude plant
material of Bacopa monnieri and quantitative determination of bacopaside I and bacoside A
by HPLC. Phytochem Anal 23:502–507. https://doi.org/10.1002/pca.2347
76. Aguiar S, Borowski T (2013) Neuropharmacological review of the nootropic herb Bacopa
monnieri. Rejuvenation Res 16:313–316
77. Eisvand F, Razavi BM, Hosseinzadeh H (2020) The effects of Ginkgo biloba on metabolic
syndrome: a review. Phytother Res 34:1798–1811
78. Chassagne F, Huang X, Lyles JT, Quave CL (2019) Validation of a 16th century traditional
Chinese medicine use of Ginkgo biloba as a topical antimicrobial. Front Microbiol 10:775.
https://doi.org/10.3389/fmicb.2019.00775
79. Okhti ZA, Abdalah ME, Hanna DB (2021) Phytochemical structure and biological
effect of Ginkgo biloba leaves: a review. Int J Pharm Res 13. https://doi.org/10.31838/
ijpr/2021.13.02.180
80. Banin RM, Machado MMF, de Andrade IS et al (2021) Ginkgo biloba extract (GbE) attenu-
ates obesity and anxious/depressive-like behaviours induced by ovariectomy. Sci Rep 11:44.
https://doi.org/10.1038/s41598-020-78528-3
81. Fang J, Wang Z, Wang P, Wang M (2020) Extraction, structure and bioactivities of the polysac-
charides from Ginkgo biloba: a review. Int J Biol Macromol 162:1897
82. Alok S, Jain SK, Verma A et al (2013) Plant profile, phytochemistry and pharmacology of
Asparagus racemosus (Shatavari): a review. Asian Pacific J Tropl Dis 3:242–251. https://doi.
org/10.1016/S2222-1808(13)60049-3
83. Sairam K, Priyambada S, Aryya NC, Goel RK (2003) Gastroduodenal ulcer protective
activity of Asparagus racemosus: an experimental, biochemical and histological study. J
Ethnopharmacol 86:1–10. https://doi.org/10.1016/S0378-8741(02)00342-2
286 R. Basu et al.
84. Pandey AK, Gupta A, Tiwari M et al (2018) Impact of stress on female reproductive health dis-
orders: possible beneficial effects of shatavari (Asparagus racemosus). Biomed Pharmacother
103:46–49
85. Pan SY, Litscher G, Gao SH et al (2014) Historical perspective of traditional indigenous
medical practices: the current renaissance and conservation of herbal resources. Evid Based
Complement Alternat Med 2014:1–20
86. Sen S, Chakraborty R, De B (2011) Challenges and opportunities in the advancement of
herbal medicine: India’s position and role in a global context. J Herb Med 1:7–75. https://doi.
org/10.1016/j.hermed.2011.11.001
87. Prakash J, Srivastava S, Ray RS et al (2017) Current status of herbal drug standards in the
Indian pharmacopoeia. Phytother Res 31:1817–1823. https://doi.org/10.1002/ptr.5933
88. Mukherjee PK, Wahile A (2006) Integrated approaches towards drug development from
Ayurveda and other Indian system of medicines. J Ethnopharmacol 103:25–35. https://doi.
org/10.1016/J.JEP.2005.09.024
89. Singh H, Chawla AS, Kapoor VK (1985) 5 medicinal chemistry research in India. Prog Med
Chem 22:243–266. https://doi.org/10.1016/S0079-6468(08)70232-7
90. Isharwal S, Gupta S (2006) Rustom Jal Vakil: his contributions to cardiology. Tex Heart Inst
J 33:161–170
91. Batiha GES, Beshbishy AM, El-Mleeh A et al (2020) Traditional uses, bioactive chemical
constituents, and pharmacological and toxicological activities of Glycyrrhiza glabra L. (faba-
ceae). Biomolecules 10:352
92. Pan Q, Pan H, Lou H et al (2013) Inhibition of the angiogenesis and growth of Aloin in human
colorectal cancer in vitro and in vivo. Cancer Cell Int 13:69–78. https://doi.org/10.1186/1475-
2867-13-69
93. Surh YJ, Park KK, Chun KS et al (1999) Anti-tumor-promoting activities of selected pungent
phenolic substances present in ginger. J Environ Pathol Toxicol Oncol 18:131–139
94. Sen S, Chakraborty R (2015) Toward the integration and advancement of herbal medicine:
a focus on traditional Indian medicine. Botanics: Targets Therapy 5:33–44. https://doi.
org/10.2147/btat.s66308
95. Anurekha J, Gupta VB (2006) Chemistry and pharmacological profile of guggul-A review.
Indian J Trad Knowl (IJTK) 05:478–483
96. Singh K (2005) Development of supply chains for medicinal plants: a case study involving
the production of vinca rosa by small farmers in the Patna District of Bihar India. … Food
Chain, …:1–16
97. Payyappallimana U (2010) Role of traditional medicine in primary health care: an overview of
perspectives and challenges. Yokohama J Soc Sci 14:57–78
98. Patwardhan B, Mashelkar RA (2009) Traditional medicine-inspired approaches to drug dis-
covery: can Ayurveda show the way forward? Drug Discov Today 14:804–811
99. Mandal SC, Mandal M (2011) Quality, safety, and efficacy of herbal products through regulatory
harmonization. Ther Innov Regul Sci 45:45–53. https://doi.org/10.1177/009286151104500105
Conservation of RET Medicinal and
Aromatic Plants, Their Traditional
Medicines and Current Practices in Indian
Himalayan Region
1 Introduction
India has been a hub of the treasure of traditional knowledge for ages which, besides
others, also explains the way outs for solving most of the present-day problems.
Plants are the primary source of all medicines worldwide, providing mankind with
new remedies. Natural sources of medicines maintain equilibrium between agricul-
ture and industrial sectors in terms of demand for natural medicine and economy,
and there is also a minimum harmful effect on human bodies. The traditional medic-
inal plant system plays a vital role in remote areas. In the primary healthcare sector,
little work so far has been done regarding the documentation and promotion of
associated knowledge [1]. Medicinal and aromatic plants are always in high demand
worldwide in both areas of health management, that is, traditional and modern sys-
tems of medicine. The traditional medicine system works through two distinct
streams, one is local, tribal or folk, and the other is codified or structured. Indian
Medicine Systems such as Ayurveda, Homoeopathy, Siddha, and Unani are formed
from both sources, plants and animals, but electrohomeopathy medicines are pre-
pared from purely herbal sources. Cultivation of medicinal and aromatic plants in
higher Himalayan ranges are new trends and is the only last option for maintaining
the wild population of medicinal plants, which are most endangered and fulfil the
industrial demands of pharmaceutical industries in global markets [2, 3]. In India
alone, less than 10% of the medicinal plants traded in the country are cultivated, and
about 90% are collected from the wild, very often in a destructive and unsustainable
manner [4].
However, in recent years, attention has been given to the development of propa-
gation methods and collection (which can support conservation objectives overall)
[5]. Most medicinal plants are being extracted from wild populations for drug and
pharmaceutical industries, traditional use and research purposes. These activities
adversely affect the existence of a number of plants, particularly those of high com-
mercial value. Further, the developmental activities in the higher Himalayan region
without rehabilitation work of these valuable resources are one of them.
1.1 ISM
India is a diverse rich country not only in terms of biodiversity but also in its unique
Traditional medicinal system and this integration leads to formation of AYUSH,
which is system of Ayurveda, Siddha, Yoga and Naturopathy. It’s an old Indian tradi-
tion medicine system and practiced in our country, multifaceted culture and tradi-
tional medicines evolved over centuries blessed with a plethora of traditional
medicines and practices. A separate department of Indian Systems of Medicine and
Homoeopathy (ISM & H) was set up in 1995 to ensure the optimal development and
propagation of AYUSH healthcare systems. The department of ISM & H was
renamed as the department of AYUSH (an acronym for – Ayurveda, Yoga and
Naturopathy, Unani, Siddha and Homoeopathy) in November 2003 [6] (Table 1).
1.2 ASM
The Ayurvedic System of Medicine is a natural Indian medicine system that origi-
nated in India more than 3000 years ago. The term Ayurveda originated from the
Sanskrit word, Ayur = life, Veda = Knowledge). This medicine system deals with
physical and mental health and covers the art of living. In the Ayurvedic system,
Table 1 Humours (vital force, OD force, bioforce) enlisted according to different alternative
medicine systems
Ayurveda Siddha Unani Homoeopathy Electrohomoeopathy
Pita (Bile) Azal Blood Syphilis Sanguine
Vayu (Wind) Vali Phlegm Sycosis Lymphatic
Kapha (Phlegm) Aiyam Yellow bile Psora Nervous
– – Black bile – Bilious
Notes: In future, it is obvious that the use of medicinal and aromatic plants will increase day by day
for fulfilling the industrial demands, hence for the preparation of any medicines in Indian Medicine
Systems (Ayurveda, Homoeopathy, Siddha, Unani, and Electrohomoeopathy) it totally depends on
plants supply. These medicines have no harmful effects on humans and animals, hence if these
MAP’s were not comes under cultivation it will lead them to face RET category due to over
exploitation from their natural habitats. So the need for developing efficient protocols of various
Agro-techniques, in vitro propagation methods and hydroponic techniques are required
Conservation of RET Medicinal and Aromatic Plants, Their Traditional Medicines… 289
diseases are cured by using drugs (phytocompound products, animal products, and
earth material products), diets, exercise, acupressure, massage, panchakarma, etc.
According to Ayurveda, it is classified into three essential elements: Vata, Pita and
Kapha, present in all living beings. Such elements originate from five elements –
Vayu (air), Tej (energy), Akash (space), Jal (water) and Earth (soil and minerals);
Space + Air = Vata, Tej+ Jal = Pita, Jal + Earth = Kapha. Diseases are caused by
such biological elements imbalance [6].
1.3 YNSM
1.4 USM
1.5 ISM
Indian Siddha System of medicine is the oldest traditional system of healing that
originated in south India. The system belongs to the Tamil civilization. The term
Siddha comes from Siddhi, which means achievement. The system is based on the
combination of ancient medicinal practices with spiritual disciplines, alchemy, and
mysticism. Six thousand species of medicinal plants were documented and pub-
lished in different research papers of ethnobotanical literature in India, among
which 750 species are generally used in the preparation of Siddha medicine from
both plant and animal kingdoms based on minerals and substances [6].
290 R. R. Kumar et al.
1.6 HMS
1.7 ESM
Electrohomeopathic system of medicine from Italy, but now the Indian government
allow these practices and research development in this system. The term of electro-
homeopathy means that Electro = Electro word comes from the electro i.e, bioen-
ergy that is extracted from medicinal plants by cohabitation methods, While
Homeopathy word means a type of medical science which treats the diseases by
using similar amount of small doses of herbal remedies as prescribed by physician.
The father of electrohomeopathic system of medicine is Count Ceaser Mattei.
According to this system of electrohomeopathy remedies that have capability to
gently stimulate the natural body’s healing system (Immune system) and also equi-
librium between the blood and lymph, hypo, hyper function physical and chemical
consistency of the cells or tissues established in a same manners as that of healthy
state of the body [7, 8].
1.8 SEM
This chapter focuses on the uses of medicinal plants. Pharmaceutical companies use
nearly 90% of medicinal plants from natural sources, which is considered to be the
main cause for the extinction of various species of medicinal and aromatic plants in
the Himalayan region. Government agencies, institutions and non-government
organizations have taken steps to cultivate medicinal plants in suitable habitats for
conservation purposes and further use in future. Here we have presented the data
based on extensive surveys done during 2018–2021 all over the Uttarakhand state in
both temperate and subalpine villages (Fig. 1), along with some research papers
and books.
Fig. 1 Glimpses of various survey and awareness meetings for collecting TMK and other techni-
cal know-how
292 R. R. Kumar et al.
3 Results
Uses of medicinal plants are also discussed in many ancient texts like Ayurveda and
in many scientific books; these literatures also focus on the importance of medicinal
herbs. Several literature that discuss the importance of herbal philosophy are
available not only in India but also in other countries like Italy, Greece and China.
Bioactive or phytochemical compounds are found in whole parts (roots, tuber, rhi-
zome, stem, leaves and flowers) of medicinal plants. Parts used from medicinal
plants basically depend upon the amount of phytochemical and specific regions of
plants as varied species to species of medicinal and aromatic plants. Roots, rhi-
zomes, tubers, as discussed in Tables 2 and 3, underground (tubers, roots and rhi-
zomes) parts are used for some species (Aconitum species, Saussurea costus,
Saussurea obvallata, Inula recemosa, Nardostachys grandiflora, Valeriana walichii,
Podophyllum hexandrum, Rheum species, Picrorhiza kurroa) to cure human and
animal related health problems. Some species (Swertia chirayita, Swertia ciliata,
Swertia speciosa, etc., of whole parts of plants, are used for curing the disease of
animals and humans. Therefore, we should always use plant parts with proper post-
harvest practices as well as Good Agricultural Practices (GAPs), that is, under-
ground parts or upper parts for the preparation of medicine.
4 Conclusion
MAPs are the spinal cord of the Indian AYUSH System since India is a rich hotspot
of many primary and secondary centres of origin, especially the Indian Himalayan
Region; hence the role of traditional medicinal knowledge is very important and
must be encouraged in a scientific, holistic manner by means of cultivation practices
so that the rich biodiversity would be maintained. Yet there is a lot of scope in eth-
nomedicinal concepts because very little scientific approach has been made related
to this field, and there is much more to be revealed, but before that, we have to
conserve these RET species of MAPs because all pharmaceutical products need to
be prepared from plant sources; hence, medicinal and aromatic plants will play a
crucial role in solving many futuristic health problems like COVID and its other
mutant variants like omicron, Alzheimer’s disease, Parkinson disease, etc.
Conservation of RET Medicinal and Aromatic Plants, Their Traditional Medicines… 293
Fig. 2 (a) Inula racemosa, (b) Saussurea costus, (c) Picrorhiza kurroa, (d) Bergiana ciliata, (e)
Valeriana wallichii, (f) Paris polyphylla, (g) Rheum emodi, (h) Rheum moorcroftianum (i)
Aconitum violaceum, (j) Aconitum balfourii, (k) Aconitum heterophyllum, (l) Swertia chirayita
(m) Swertia ciliata, (n) Swertia speciosa, (o) Saussurea obvallata, (p) Nardostachys grandiflora,
(q) Malaxis muscifera, (r) Dactylorhiza hatagirea, (s) Angelica glauca, (t) Podophyllum
hexandrum
302 R. R. Kumar et al.
Fig. 2 (continued)
Conservation of RET Medicinal and Aromatic Plants, Their Traditional Medicines… 303
Fig. 2 (continued)
304 R. R. Kumar et al.
Fig. 2 (continued)
References
1. Gidey M, Asfaw Z, Woldu Z (2009) Medicinal plants of the meinit ethic group of Ethopia: an
ethnobotanical study. J Ethnopharmacol 5570:9
2. Dobhal P, Purohit VK, Chauhan J (2021) High frequency plant regeneration from fully mature
shoot portion of Nardostachys Grandiflora DC. Int J Conserv Sci 12:1053–1060
3. Purohit VK, Mengwal BS, Dobhal P, Chauhan J (2021) Cultivation of high altitude medicinal
and aromatic plants: a key for sustainable development and bioresource conservation in Higher
Himalayan Region (HHR) of Uttarakhand, India. Ed. Deb CR, Paul A. Mittal Publications
Daryaganj, New Delhi, pp 51–73
4. Natesh S (2000) Biotechnology in the conservation of medicinal and aromatic plants. In:
Chandha KL, Ravindran PN, Sajiram L (eds) Biotechnology in horticultural and plantation
crops. Malhotra Publishing House, New Delhi, pp 548–559
5. Rajasekharan PE, Ganeshan S (2002) Conservation of medicinal plant biodiversity in Indian
perspective. J Med Aromat Plant Sci 24:132–147
6. Ministry of AYUSH, CCRS, Government of India, New Delhi (2019) ISBN -978-81-937426-3-1
7. Kundu D (2019) The OD force. Int J Herb Plant Med 3:55. https://doi.org/10.5580/
IJHPM.54807
8. Pandey S, Pandey PS, Mohanty D (2019) Electrohomeopathy: a revolutionary system of plats
medicine. European J Biomed Pharm Sci 6:301–305
9. Parkash R (2015) Medicinal plants used by tribal communities: a study of Uttarakhand
Himalaya region. Int J Humunit Soc Sci Invit 4:55–61
10. Samant SS, Nandi KS (2009) Conservation status and cultivation of selected medicinal plants
in the Indian Himalayan region. In: Advances in agriculture environment and health. Satish
Serial Publication House, Azadpur
11. Khare CP (2007) Indian medicinal plants. In: An ilustrated dictionary, Springer-Verlag, Berlin/
Heidelberg, ISBN 978–0–387-70637-5
12. Nautiyal MC, Nautiyal BP (2004) Agrotechniques for high altitude medicinal and aromatic
plants. Bishen singh Mahendra pal singh, Publisher and Distributors of scientific books, ISBN:
81-211-0338-X
13. Chadha KL, Ravindran PN, Sahajram L, Natesh S (2000) Biotechnology in the conservation of
medicinal and aromatic plants. In: Chadha KL, Ravindran PN, Sahajram L (eds) Biotechnology
in horticulture and plantation crops. Malhotra Publishing House, New Delhi, pp 548–561
14. Madhu KS, Phoboo S, Jha KP (2010) Ecological study of Paris polyphylla ecological society
Nepal. Ecoprint 17:87–93
Conservation of RET Medicinal and Aromatic Plants, Their Traditional Medicines… 305
15. Malik AM, Bhatt AS, Fatima B, Ahmad BS, Sidiqui S, Shrivastava P (2016) Rheum emodi as
valuable medicinal plant. Int Acad Sci Eng Technol 5:35–44
16. Shyaula LS (2011) Phytochemicals, traditional uses and processing of Aconitum species in
Nepal. Nepal J Sci Technol 12:171–178
17. Pelletier SW, Aneja R, Gopinath KW (1968) The alkaloids of Aconitum heterophyllum wall:
isolation and characterization. Phytochemistry 7:625–635
18. Pelletier SW, Ateya AM, Finer-Moore J, Mody NV, Schramm LC (1982) Atisenol, a new enta-
tisene diterpenoid lactone from Aconitum heterophyllum. J Nat Prod 45:779–781
19. Aneja R, Locke DM, Pelletier SW (1973) The diterpene alkaloids: the structure and stereo-
chemistry of heteratisine. Tetrahedron 29:3297–3308
20. Kant R (2015) Survival threats and conservation of Malaxis muscifera (Lindl). Kuntze, a
threatened medicinal Orchid at Fagu, Himachal Pradesh. IJBASA 1:20
21. Chauhan SR, Dutt P (2015) Swertia speciosa wall: a new source of amaroswerin. Med
Plants 7:2
22. Kumar RR, Chandola V, Chauhan J, Prasad P, Purohit VK (2009) Medicinal importance of
Swertia speciosa. Wall.ex.D.Don. J Emerg Technol Innov Res 6:6
Traditional Amchi Medicinal Practice
in Trans-Himalaya of Nepal: Conservation
and Bioprospecting
1 Introduction
N. Chaudhary (*)
Central Department of Botany, Tribhuvan University, Kirtipur, Kathmandu, Nepal
Resources Himalaya Foundation, Sanepa, Lalitpur, Nepal
S. K. Ghimire
Central Department of Botany, Tribhuvan University, Kirtipur, Kathmandu, Nepal
R. P. Chaudhary
Research Centre for Applied Science and Technology (ReCAST), Tribhuvan University,
Kirtipur, Kathmandu, Nepal
medications [5]. This system of medicine plays a pivotal role in health care in many
remote villages in the Himalaya. In addition, it also emphasizes water and spring
cleanliness, a good diet, and a healthy way of living [6].
The highlands of the Himalaya, the Tibetan mountain range, and its adjoining
areas are among the few remaining areas where the Amchi system of medicine
(Sowa Rigpa) remains effective [7]. This medicinal practice is extended northwards
to Mongolia and beyond, eastwards to Sichuan and Yunnan, westwards to Ladakh,
and Himachal Pradesh (north-western India) and a few parts of the former Soviet
Union, and southern regions to Nepal trans-Himalaya [1, 6]. It is the most legitimate
and popular medical system in the mountainous areas of Nepal adjoining the Tibetan
Plateau [2]. However, amchi in the Himalaya are now facing several challenges,
mostly related to the unavailability and increased cost of ingredients used in com-
pounding medications, financial constraints, lack of formal state recognition, and
loss of traditional knowledge [3, 8]. Especially, India, Nepal, and China have been
facing the loss of traditional knowledge along with the loss of species [1, 9–12].
Overexploitation of medicinal plants in response to the commercial demand from
natural product industries in the region and beyond has increased the risk of extinc-
tion of many valuable species [13]. Therefore, critical systematic review and
research is necessary on the traditional amchi practice and the status of medicinal
species. The objective of the review is to discuss about the amchi’s medicinal prac-
tice and the major issues related to this system in the trans-Himalaya of Nepal.
For a systematic review, we searched accessible literature related to Sowa Rigpa.
We used “Google Scholar” for the web-based search using keyword combinations
“Nepal,” “India,” “China,” “Tibet,” “Myanmar,” “Bhutan,” “Ladakh,” “Himachal
Pradesh,” “Himalaya,” “Trans-Himalaya;” and “amchi,” “Sowa Rigpa,” “Tibetan
traditional medicine.” A total of 92 literature were retrieved but most of them
described the use of plant species, where 12 of the literature were not able to
be downloaded. We downloaded 47 related articles and reports through the initial
screening. In addition, we used our own experience and observation while conduct-
ing field work in different parts of Nepal.
Traditionally, amchi’s knowledge and practice have been passed down within a fam-
ily as a lineage-based tradition or from teacher to disciples under the guru–shisya
tradition. Most of the amchi in Nepal (mainly those in Dolpa and Mustang Districts)
are the sixth generation of an unbroken family lineage [3]. Medical knowledge is
also taught in the monastery as part of religious training. Some amchis in Nepal
have also obtained higher Sowa Rigpa degrees from formal Tibetan medical schools
in India or Tibetan Autonomous Region of China. Normally, amchis begin to treat
patients after at least five to seven years of study and practice as an assistant. Most
amchis in Nepal, except those in urban areas, provide health care services almost free
of cost.
Traditional Amchi Medicinal Practice in Trans-Himalaya of Nepal: Conservation… 309
According to Sowa Rigpa philosophy, a healthy mind and a healthy body are con-
nected, and health or illness is related to our relationship to attachment, anger, and
ignorance. Wind (rlung), bile (mkhris pa), and phlegm (bad kan) are the three dynam-
ics (humors), the imbalance of which leads to illness. In addition, earth, water, fire,
air, and space/consciousness are the five elements that help to regulate our health.
Amchis perform their diagnosis of disease by examining a person’s overall physical
condition. The following are the common approaches used for disease diagnosis [2,
3, 10, 14]: interviewing, visual examination, pulse analysis (tactile test), and urine
examination. First, the patient’s medical and social history, diet, and behavior are
carefully considered to find out the causative factor, the site of illness, and the signs
and symptoms. Then, patient’s physical characteristics and complexion of five sen-
sory organs (eyes, ears, nose, tongue, and skin) are visually examined. Pulse exami-
nation is the most important component of diagnosis, which involves the analysis of
the relationships between the three dynamics (wind, bile, and phlegm), the five ele-
ments and the seven bodily constituents. The other areas of concern include body
temperature and inflammations. Finally, a thorough examination of urine is done,
which includes the use of sight, smell, taste, and touch. Pulse analysis and urine
examination form the most distinctive and important parts of diagnosis.
Amchis apply a variety of approaches for the treatment of illnesses [2, 3, 10, 14];
the most important ones are diet, modification of behaviors, use of herbal medi-
cines, and physical therapy. In Sowa Rigpa, the first recommendation for the treat-
ment of disease constitutes diet and behavior practices. The behavioral approach
focuses on routine, seasonal, and incidental behaviors. When the diet and routine
behavior are not effective in relieving the disease conditions, then medicines as well
as physical therapy are prescribed. The medicines are prepared from natural
resources, including plants, minerals, and animal-derived products and adminis-
tered in different forms such as decoctions, pills, powder, gruel (thin porridge),
medicinal butter, medicinal calx (ash powder), concentrated extractions, medicinal
incense, medicinal wine, or other formulas. Apart from prescribing natural drugs,
the physicians may apply other therapeutic techniques such as massages, hot and
cold compresses, mineral spring bath therapy, medicinal baths, golden needle ther-
apy, bloodletting, and moxibustion (heat therapy by burning the leaves of moxa
herbs, which include several species of Artemisia, Ajania, Anaphalis, and
Leontopodium). Generally, surgery is no longer practiced by amchi; some minor
operations are done including the draining of abscesses.
Amchis have been serving local communities in the remote mountainous areas of
Nepal since ancient times. Even today, in many of the remote high-altitude areas,
Amchi medicine is the only form of health-care service available. The modern allo-
pathic medicine with its less formal infrastructure has brought few benefits or is
virtually non-existent in most of the remote mountainous areas of Nepal. Therefore,
310 N. Chaudhary et al.
in such areas, amchi’s service has been indispensable for maintaining people’s
health. In addition to the health care services, the contribution of amchi from the
Himalayan region of Nepal and particularly those from Dolpo (present-day Dolpa
District) for the advancement of Sowa Rigpa theory and practice has been well
known [3, 15]. Now, this practice is under threat in Nepal as it faces several chal-
lenges of socio-political, economic, and ecological in nature, mostly related to the
lack of formal recognition, and inappropriate support from the governmental and
non-governmental sectors, loss of traditional knowledge, loss of biodiversity and
unavailability and increased cost of the ingredients, and lack of a mechanism for
sharing and integrating amchi’s knowledge and practices [2, 3].
In most of the Asian countries, including Bhutan, China, India, and Mongolia,
where Sowa Rigpa is practiced, this system has been recognized officially. In
Bhutan, Sowa Rigpa has been officially recognized as a Bhutanese Traditional
Medicine and it has been integrated in the state healthcare system. In India, it was
officially recognized as an Indian System of Medicine. In Nepal, despite more than
20 years of effort from the amchi community, Sowa Rigpa still lack full support and
recognition from the Government of Nepal [3]. Recently, the Public Health Service
Act, 2075 (2018), of the Government of Nepal has just listed Sowa Rigpa under the
alternative medicine but does not provide sufficient space to recognize it as a Nepali
System of Medicine. In order to help protect the traditional knowledge and practice
of amchi and provide efficient health services for remote high-altitude communi-
ties, Sowa Rigpa need full official recognition, and the profession of amchi should
be properly institutionalized with a full certification and registration system. Sowa
Rigpa should be supported through universities, hospitals, pharmacies, and depart-
ments within health and education ministries of the Government of Nepal [3].
In the trans-Himalaya of Nepal, amchis pass on their knowledge and skills
mostly from father to son/daughter or apprentices. However, only a few persons
take interest in Amchi medicine. Since amchi’s knowledge and profession are
declining, the standard of health care in the region is likely to suffer. The loss of
amchi’s knowledge can be attributed mostly to the lack of access to resources, lack
of infrastructure, and human resources and monetary problems. The traditional way
of collecting herbs for the preparation of medicines is labor-intensive and difficult
due to the migration of skilled human resources to the cities [3].
The most important challenge to sustain this practice in the remote trans-
Himalayan region of Nepal is the unavailability and increased cost of several ingre-
dients traditionally being used in compounding medication [3]. Many amchi feel
difficulty in acquiring their full skills due to declining population or lack of proper
medicinal plants. Recent study by Ghimire et al. [3] has shown that amchi in Nepal
utilize ingredients from over 570 species of plants and 54 different types of minerals
(extracted from stones, gems, precious metals, and soil) for compounding medica-
tions. Of the plant-based medicines, over 100 species are locally rare, including
about 40 species which are either threatened and included in IUCN Red List or are
regulated by CITES. These also include medicinal plant species legally protected by
the Government of Nepal but are of great significance for the traditional Amchi
medicinal practice. Commercial overharvesting has been identified as the main
Traditional Amchi Medicinal Practice in Trans-Himalaya of Nepal: Conservation… 311
threat for the viability of medicinal plant populations in the high Himalaya of Nepal,
although habitat destruction, fragmentation and loss are also important. Harvesting
of plants for traditional use by amchi themselves does not necessarily pose a serious
ecological threat as they need very small quantity, usually 0.1–8 kg per amchi per
year (depending on the species and types of formulations) in the Nepal’s trans-
Himalaya [3], and they use unique cultural criteria for the selection of medicinal
plants and apply harvesting methods, following the guidelines indicated in the stan-
dard Sowa Rigpa texts, which are often evaluated as sustainable [3, 15, 16].
Commercial harvesting has imposed extreme pressures on the populations of sev-
eral valuable species including Aconitum spp., Ophiocordyceps sinensis,
Neopicrorhiza scrophulariiflora, Dactylorhiza hatagirea, Nardostachys jatamansi,
Fritillaria cirrhosa, Valeriana spp., Dendrobium spp., so that these are no longer
available to most of the trans-Himalayan amchis.
In addition, ingredients from several exotic plant species (26 of such species)
have been documented recently [3], including Aquilaria malaccensis, A. sinensis,
Aucklandia costus, Commiphora wightii, and Pterocarpus santalinus, highly needed
for Amchi Medicine are now not easily available as these are threatened and included
under IUCN Red List or regulated by CITES. Price has dramatically increased for
such plant-based ingredients, which most amchi are unable to pay given their very
low annual income. The professional ethics of amchi is based on Buddhist and Bon
concepts of universal compassion and do not allow them to charge their patients
fees for their service. But in the present context, amchi cannot afford to purchase
plants and other products from the lowlands [3, 15]. Only a few amchi can place an
order and get sufficient plant materials and products supplied, whereas the majority
do visit the lowlands to sell medicinal herbs collected in their localities during win-
ter and to buy materials for ingredients. Similarly, most of the amchi also cannot
afford to purchase expensive minerals, precious metals, and gems due to high price.
Earlier, amchi also used wildlife parts such as musk pod, gall bladder of the
Himalayan black bear, rhino horn, etc., to prepare quality medicine; however, these
are endangered species and the GoN has put a ban on poaching of endangered wild
animals. Therefore, most learned amchis in Nepal practice plant- or mineral-based
substitute of animal parts [3].
Despite recent efforts in documenting amchis knowledge and practices from
trans-Himalayan region of Nepal, there is still a need to assess the availability and
ecological status of species used in Sowa Rigpa. Particularly, the use of rare and
threatened species in medicine has posed ethical and environmental challenges to
amchi. Therefore, eminent amchis have identified locally available common plants
and minerals as possible substitutes for rare and threatened plant and animal parts
used in the preparation of medicine [3, 15].
Thousands of substances are recorded as medicinal in Sowa Rigpa practiced in
Asia [17], and there are regional and local variations in the selection of these
resources. In many cases, the use of enormous resources is yet uncertain. There is
also a lack of mechanism for integrating knowledge and practices covering amchis
from all over Nepal and neighboring countries.
312 N. Chaudhary et al.
The selection of plant species for medicine is guided by cultural practice, which is
directly related to the question of efficacy.
(i) Compound medicine Traditional Amchi medicinal practice is frequently com-
posed of more than one constituent. Compound medicines can be pharmacologi-
cally different from their individual plant constituents. In plant combinations, the
activities of two or more plants may add to increase efficacy such as (i) the action of
one constituent may be potentiated by another so that the effect is greater than the
additives themselves; (ii) one constituent may diminish the activity of another
through antagonistic interaction; or (iii) the effect may be synergistic, when one
constituent substantially amplifies the activity of another [21]. For example, plants
used as common additives, such as black pepper (Piper nigrum), long pepper (Piper
longum), and ginger (Zingiber officinalis), potentiate the action of additional plants
in the medicine by markedly increasing the bioactivity of their active constituents
[22]. The association of plants may diminish the toxicity of their individual con-
stituents (such as tannins and saponins) [23]. The Amchis in Dolpo use Nepal aco-
nite (Aconitum spicatum), a highly poisonous plant, for treating coughs, bile fever,
pulmonary and intestinal infections, headaches, cuts and wounds after detoxifying
the root tuber by boiling it with an extract of Terminalia chebula [15]. Amchis also
purchase medicinal plant resources from the lowlands Tarai that include
Cinnamomum zeylanicum, Phyllanthus emblica, Santalum album, Terminalia bel-
lirica, and Terminalia chebula, and these are mixed to make drugs with locally
314 N. Chaudhary et al.
available plants. In Nepal, the medicinal plants that are being used in treating
common ailments and diseases need authentication and standardization of efficacy
and dosage with the help of pharmacognostic analyses in the laboratory so as to
encourage the indigenous peoples’ and local communities (IPLCs) to continue to
use the effective medicinal plants and discourage the use of ineffective and poten-
tially harmful ones.
(ii) Harvesting sustainability Amchis usually begin their studies early in their
teenage years. Apart from learning the diagnosis of illnesses/diseases and therapeu-
tic measures, they are trained to identify the high-altitude plants in their natural
setting during summer, and low-altitude plants during winter. In addition, they also
learn the related aspects of biology, distribution, habitat, density and local availabil-
ity, as well as sustainable harvesting and cultivation practices. Amchis possess an
in-depth knowledge of how to harvest plants at the proper stage for better medicinal
efficacy and sustainability. In the trans-Himalayas, such as upper part of Manang
District, harvesting is managed culturally, with the head lama restricting the har-
vesting of both fodder grass and important medicinal plants to a specific period
(September–October) just before crop harvesting begins, so that annual and biennial
plants can complete their full life cycle [24].
The amchis possess in-depth knowledge about the ecology and economy of plant
species and are quite familiar with which species are becoming difficult to find, due
to either limited geographical distribution or habitat destruction or overexploitation.
They have a practical interest in the conservation of medicinal plants, which is valu-
able for developing management guidelines for plant conservation, as bulk of
medicinal resources is derived from plants (Box 2).
The trade in medicinal plants from Nepal to Tibet dates back to the eighth century
(728 CE/AD) and constitutes a considerable part of Tibetan medicine’s materia
medica [26]. Some vulnerable plant species that are carefully collected by the amchi
in Nepal for traditional medicinal practice include Aconitum naviculare (bongkar),
Aconitum spicatum (bongnak), Arnebia benthamii (muktsi), Corallodiscus lanugi-
nosus, Cypripedium himalaicum (dakya habo), Dactylorhiza hatagirea (wanglag),
Halenia elliptica (tikta), Meconopsis grandis (upal ngon po), Meconopsis horridula
(ajaktsergun), Nardostachys grandiflora (pangpoe), Neopicrorhiza scrophulariiflora
(honglen), Rheum australe (churtsa), and Valeriana jatamansii (nahpoe) [15].
In general, the collection of medicinal plants for traditional local use is not a
problem, since this use has usually developed gradually, and in harmony with nearby
natural ecosystems. However, when selected species are gathered in large quantities
and supplied to meet increasing demand of national and international trade, then
pressure can quickly mount and cases of overexploitation are common [27].
It is obvious that medicinal plant species collected for commercial purposes rep-
resent the most popular and often most effective herbal remedies. The majority of
species that were popular in the past are still popular today. It is interesting to note
that the widespread commercial harvesting and sale of the same plants have been
going on for a long time in Nepal [28]. Commercial collectors of medicinal plants
are those people whose main aim is not resource management but earning money.
To obtain a clear view of the relationship between people and their natural environ-
ment and to suggest sustainable options, it is necessary to gather data, such as the
type, source, and quantity of the resources used, their importance within the cultural
context and alternatives if the resource is scarce and needs to be conserved for the
preservation of genetic diversity [29]. The knowledge possessed by the Dolpo
Amchis relating to the use and management of medicinal plants will contribute
towards devising appropriate systems for sustainable harvesting, which may ulti-
mately be transferred to commercial collectors who tend to overexploit the resources.
In trans-Himalaya (Dolpo, Mustang and Manang), amchis and women have a keen
interest in the conservation and management of medicinal plants [15, 30]. In terms
of conservation, climate change possess significant challenges for highly traded pri-
oritized medicinal plants [31]. It is experiencing the alternation of the habitat, dis-
tribution, ecology, and phenology of the medicinal plants in the Himalaya. As a
result, there is an adverse effect on medicinal plants, and some of the species may
get extinct too. These changes are likely to have negative consequences on the vul-
nerable population across the Himalaya [32].
7 Conservation Approach
(i) Education Educational program for the young generation is an important way
of practicing medicinal practice. Supporting programs encourage children to study
the local uses of plants. This approach would ensure that the ancient lore of the
Himalayan plants will be protected within the communities and customary institu-
tions. This will raise awareness among the people that conservation education can
be a powerful means of preserving both their culture and nature [33]. In the educa-
tional program, attention should be given for the diversity of in-depth knowledge
within and cultural groups for the resource management and developing scientific
understanding of the ecological status of key resources [16].
(ii) Storage of medicinal plants Medicinal plants and their voucher specimens
(herbarium specimens) stored in the community center or school provide local peo-
ple with a permanent record of the useful species documented in the course of sus-
tainable use and conservation. It is important that the materials are stored in tightly
sealed cabinets protected from humidity and insects since the biological activity of
plant materials ceases after a few years of storage. An antibacterial and antifungal
study of a total of 19 medicinal plants in Nepal was conducted by Griggs et al. [34]
showing that after six years of storage, 3 ceased all activity, 6 retained all activity,
and the remaining 10 plants retained only partial activity. This type of study is
extremely important for assessing the “self-life” of herbal medicines which are
being utilized by the communities, herbal practitioners and amchi in Nepal and
provides a reason to test the efficacy of stored medicinal plants used by Amchis and
pharmaceutical companies.
(iii) Nature tourism In the Himalaya, nature tourism is based on spectacular land-
scapes, and rich and unique biodiversity combining with the context of a specific
cultural heritage – all interacted in sustainable manner. Nature tourism in Nepal is
largely confined to the activities of mountaineering and hiking, so that a sustainable
nature tourism that focuses on natural history, customary practices and culture is yet
to be explored and established [35].
Ethnoecologists can play a key role in designing interpretive programs for tour-
ists or other visitors that enhance the conservation of traditional ecological knowl-
edge. Naturalists in Kinabalu Park (a 735 km2 protected area in Sabah, Malaysia)
have been training local Dusum guides who are knowledgeable about the plants and
animals and speak Western and local languages. The bilingual Dusum guides
accompany the tourists, explaining the use of plants such as rattan palms and other
forest products both in the natural history museum located at the park headquarters,
and along the summit trails [33]. The prospects for such a program in the Himalaya
have yet to be adequately explored, strengthened, and popularized.
Traditional Amchi Medicinal Practice in Trans-Himalaya of Nepal: Conservation… 317
8 Bioprospecting
(ii) Searching plants for new medicine One of the crucial questions is how we
should begin the search for new medicines with the vast number of plants to be
studied. Cox and Balick [42] have discussed three approaches. The first approach is
random screening; one can simply screen everything that can be collected in quan-
tity, the so-called mass or blind screening approach. Obviously, such a strategy
requires investment of time and money. The discovery of taxol from Taxus brevifo-
lia (the Pacific yew tree) is an example of a random-screening program conducted
by the National Cancer Institute (NCI). A second approach is the chemotaxonomy-
oriented approach, which is more guided in that it screens those species that belong
to certain families or genera and are likely to contain certain classes of natural
compounds, such as alkaloids, steroids, terpenoids, and amino acids. A realistic
ethnopharmacological approach selects those plants which are being used for rem-
edies/medicines by the indigenous peoples and local communities for further study.
This approach calls for the exploration of biologically active agents, traditionally
employed or observed by traditional healers and indigenous communities.
A number of essential drugs used in allopathic medical practice are derived from
plants, and have been developed based on indigenous knowledge; a few examples
include aspirin from Filipendula ulmaria, codeine from Papaver somniferum, ipe-
cac from Psychotria ipecacuanha, pilocarpine from Pilocarpus jaborandi, pseudo-
ephedrine from Ephedra sinica, quinine from Cinchona pubescens, reserpine from
Rauvolfia serpentina, scopolamine from Datura stramonium, theophylline from
Camellia sinensis, and vinblastine from Catharanthus roseus [42].
Only 1.5% (about 4000 species) of the flowering plants of the world have been
screened for pharmaceutical compounds; and of this number, 3% currently provide
major drugs. Lower groups of plants, animals and microorganisms remain neglected
[43]. Some promising taxa that have yielded chemicals for major drug development
include Aesculus hippocastanum (horse chestnut), Fraxinus rhynchophylla, Camellia
sinensis (tea), Ephedra sinica (ma-hang), Podophyllum peltatum (may apple),
Hyoscyamus niger (henbane), Rhododendron molle (yellow azalea), Artemisia mari-
tima (levant wormseed), Corydalis ambigua (birthwort), Thymus vulgaris (common
thyme), Valeriana officinale (valerine), Justicia adhatoda (malabar nut), Daphne gen-
kwa (pinyin; yuan hua). These taxa indicated a positive correlation between tradi-
tional medical use of the plant and the current therapeutic use of the chemical extracted
from the plant [43]. The other species of the above genera found in the Himalayan
region could provide valuable information under the chemotaxonomy- oriented
approach, while the knowledge of the Amchis and indigenous peoples and local com-
munities in the Himalaya lend themselves to the ethnopharmacology approach.
and the Nagoya Protocol on “access to genetic resources and fair and equitable shar-
ing of benefits arising from their utilization” adopted in 2010 [44]; however, there is
a continuing controversy over the impact of intellectual property rights (IPRs) sys-
tems on the traditional knowledge of indigenous peoples and local communities.
Many indigenous peoples’ representatives and outside commentators feel that exist-
ing IPR systems are inadequate for protecting indigenous intellectual and cultural
property rights (e.g., Mataatua Declaration [45]).
The UN Convention on Biological Diversity, particularly Article 8(j), addresses
traditional knowledge and equitable benefit sharing. The vital role of indigenous
peoples and local communities [including women] is focused in its call to “respect,
preserve, and maintain knowledge, innovations and practices of indigenous and
local communities embodying traditional lifestyles relevant for the conservation
and sustainable use of biological diversity and promote their wider application with
the approval and involvement of the holders of such knowledge, innovations and
practices and encourage the equitable sharing of the benefits arising from the utili-
zation of such knowledge, innovations and practices.” The article, however, does not
provide details on how to recognize, reward, and protect the contribution of local
communities, farmers, indigenous peoples, and women. The biggest challenge fac-
ing governments and society today is how to distribute the economic benefits of
biodiversity fairly and equitably. Because establishing a system of intellectual prop-
erty rights to biological resources has proved contentious. Biological diversity (both
wild and cultivated) is most prominent in developing countries, and the economic
benefits it generates are disproportionately captured by industrial nations at the
global level.
The Constitution of Nepal recognizes the importance of biodiversity conserva-
tion by incorporating different clauses on natural resource conservation (Article
51), intellectual property rights (IPRs) (Article 25) under different sections includ-
ing Fundamental Rights and Duties (Part 3), Directive Principles (Part 4), and other
sections [46]. The constitution also guarantees the rights of every person to live in a
clean environment as a fundamental right (Article 30). Article 51 of the Constitution
of Nepal requires all three tiers of the governments to protect, promote, and use
available natural resources of the country in agreement with national interest.
Further, it also adopts the concept of inter-generational equity and make equitable
distribution of benefits according to priority and preferential right to the local com-
munities (Article 51, Clause G) [47]; however, conservation of biological resources
for peoples’ livelihood, access to genetic resources and benefit sharing giving prior-
ity to the indigenous communities have yet to be addressed as Fundamental Rights
in the Constitution of Nepal.
Intellectual property is the human mind’s creation of ideas, information, and
knowledge that can be incorporated into creative or inventive works, such as inven-
tions, designs, trademarks, books, paintings, or other literary and artistic works.
Intellectual property rights (IPRs) provide legal rights to grant ownership to inven-
tors, who may be individuals, enterprises or other entities that create products by
intellectual effort. However, IPR protection is being developed to benefit the coun-
tries/companies in the developed countries that have access to high technology;
320 N. Chaudhary et al.
whereas the traditional healers, farmers, and indigenous communities in the devel-
oping countries are not given due recognition for their contribution. Therefore, the
most suitable approach for Nepal appears to be to define the role of indigenous
peoples and local communities, farmers, and their knowledge in the national legisla-
tion that brings the convention into effect [48].
Traditional knowledge is a valuable heritage for the communities and cultures
that develop and maintain it. Medicinal and food plant genetic resources represent
the most important category of all biological resources because of their importance
in primary health care and pharmaceutical products. Traditional knowledge and bio-
diversity conservation are complementary phenomena essential to human develop-
ment. Very little of the traditional knowledge has been recorded; yet, it represents an
immensely valuable source of information [49]. The indigenous knowledge of tra-
ditional healers and women, particularly in the Nepalese context, has long been
ignored. Today, however, a growing number of Nepalese researchers and institu-
tions, and international development agencies are recognizing that the traditional
knowledge, innovations and practices of indigenous peoples, and local communities
relevant to conservation and the sustainable use of biodiversity are under threat
[15, 28].
Nepal has made an attempt to incorporate such obligation as far as possible into
“Access to Genetic Resources and Benefit Sharing (AGRBS) Bill” awaiting for
approval from the parliament as national legislation. However, access to genetic
resources and benefit sharing (AGRBS) will remain incomplete unless the social
and economic well-being of people and governance issues related to the same are
addressed properly.
In this context, the Center for International Environment Law (CIEL) has pro-
posed several initiatives as the next steps in using intellectual property as a tool for
conserving traditional knowledge and biodiversity [50].
(i) Free Prior informed consent (FPIC) and mutually agreed terms (MAT) The
Nagoya Protocol ensures that the genetic resources and associated traditional
knowledge are accessed in accordance with the domestic legislative framework
meeting the provisions of Free and Prior Informed Consent (FPIC) and Mutually
Agreed Terms (MAT). Following CBD (Article 15.5), developing countries have
extended their demand to obtaining free prior informed consent (FPIC) from the
concerned country of origin before patent applications are filed. Developing coun-
tries are also demanding an amendment to Article 29 of TRIPS (The Agreement on
Trade-Related Aspects of Intellectual Property Rights) “Conditions on Patent
Application” that would be constraining patent applicants to make adequate
disclosure of the “country of origin” of the biological resources or traditional knowl-
edge. It is obvious that without an amendment to TRIPS, there is a weak legal basis
for benefit sharing.
The FPIC of all peoples and their communities must be obtained before any
research is undertaken [51]. Indigenous peoples, traditional societies, and local
communities have the right to obtain information about any program, project or
study that affects them. It is generally presumed that all potentially affected com-
munities will be provided with complete information regarding the purpose and
Traditional Amchi Medicinal Practice in Trans-Himalaya of Nepal: Conservation… 321
nature of the research activities and the probable results through FPIC and
MAT. These will also help to reasonably foresee benefits and risks of harm (be they
tangible or intangible) to the affected communities.
Key elements of FPIC and MAT, and their application procedures have been
discussed by Oli and Dhakal [52]. In order to give full force to the FPIC and MAT
requirement, the Government of Nepal established an authority under the Ministry
of Forests and Environment (MoFE) at the national level as a focal point for coordi-
nating and implementing access to genetic resource agreements.
(ii) National registers of traditional knowledge Biodiversity cannot be protected
by regulations only; the active support and participation of IPLCs are fundamental.
There is a need to address various aspects of biodiversity use and conservation at the
community level. Our knowledge of Nepal’s biological resources and associated
traditional ecological knowledge need to be completed and consolidated.
Biodiversity registration involves a systematic documentation of the resources,
knowledge, and skills of the people at the local and national level by initiation of the
people themselves. The basic philosophy behind this endeavor is that it will rejuve-
nate both the ecological basis of the resources and the social, cultural, and economic
conditions of the people by making the people knowledgeable about, capable of,
and responsible for biodiversity management.
The National Biodiversity Registration (NBR) initiated in Nepal is the beginning
of a process of creating mechanisms to (i) ensure recognition of the indigenous
knowledge of communities, (ii) avoid the misappropriation of specialized varieties
and genetic resources used by farmers, and (iii) enhance future benefit sharing [53–
55]. The Ministry of Forests and Environment has been pursuing to extend biodiver-
sity registration at province and local levels throughout Nepal. Developing a
common agreement on the registration of biological resources and knowledge
among the countries in the Hindu Kush Himalaya has yet to be worked out. For
instance, the members of the Andes Pact (Bolivia, Columbia, Ecuador, Peru, and
Venezuela) have developed a common agreement on biosafety and a special regime
for the safeguarding of traditional knowledge [56].
(iii) Document case studies on sharing of benefits from specific uses Discussions
of the impact of IPRs on the sharing of benefits from the commercial use of tradi-
tional knowledge and associated genetic resources could benefit from more fact-
based analysis of specific cases.
Traditional knowledge is usually accessed by companies through internet and
literature and, in some cases, through multiple intermediaries and reports. Rarely
companies provide grants to communities possessing traditional knowledge, with
which they work. Aveda Corporation, Minnesota, has developed production and
marketing of Bixa orellana, a common and widespread species throughout the neo-
tropics and in the “public domain” with the Yawanawa community of Brazil. Aveda
Corporation uses bixa as a colorant in lipstick and other personal care and cosmetics
products. In 1993, the partnership between Yawanawa and Aveda involved a pack-
age of benefits over the number of years that includes the supply of technical assis-
tance, start-up funds to begin initial production, local processing, transportation and
322 N. Chaudhary et al.
other logistics, facilitation of organic certification, guaranteed market for the prod-
uct; as well as distribution of benefits [57].
However, the following are some common examples of patents in which the
contribution of traditional knowledge has been ignored (Box 3).
America. Rural women are often the most knowledgeable about the patterns and
uses of local biodiversity (Box 4). Therefore, the important role of women in the
management of biological resources must be recognized, and they must be allowed
to participate in decision-making. Agenda 21 calls for women to be fully involved
in decision making and in the implementation of sustainable development activities.
In the Himalaya, for example, rural women normally play a major role in the collec-
tion of various forest products, including food and medicinal plants; in farming and
managing water resources. Hence, women’s knowledge needs to be recognized and
their participation at three levels of management be encouraged: national policy, the
meso-level and grassroots [59].
11 Conclusions
References
32. Aryal P (2015) Climate change climate change and its impact on medicinal and aromatic
plants: a review. Clim Chang 1(1):49–53
33. Martin GJ (1995) Ethnobotany: a methods manual. Chapman & Hall, London
34. Griggs JK, Manandhar NP, Towers GHN, Taylor RSL (2001) The effect of storage on the bio-
logical activity of medicinal plants from Nepal. J Ethnopharmacol 77:247–252
35. Chaudhary RP (2001) Ecotourism: bridge between biodiversity conservation and develop-
ment in Nepal. In: Watanabe T, Sicroff T, Khanal NR, Gautam MP (eds) Proceedings of the
international symposium on the himalayan environments: mountain sciences and ecotourism/
biodiversity. Hokkaido University/Tribhuvan University, Japan/Nepal, pp 31–39
36. Cushnie TPT, Cushnie B, Echeverría J, Fowsantear W, Thammawat S, Dodgson JLA et al
(2020) Bioprospecting for antibacterial drugs: a multidisciplinary perspective on natural prod-
uct source material, bioassay selection and avoidable pitfalls. Pharm Res 37(7):1–24
37. Svarstad H, Dhillion SS (2000) Responding to bioprospecting: rejection or regulation? In:
Svarstad H, Dhillion SS (eds) Bioprospecting from -biodiversity in the South to Medicines in
the North, pp 9–15
38. Pushpangadan P (2018) Traditional medicine & clinical naturopathy biodiversity, bioprospect-
ing, traditional knowledge, sustainable development and value added products: a review. Tradit
Med Clin Naturop 7(1):1–7
39. Chaudhary RP (2000a) Biodiversity prospecting in Nepal-constraints and opportunities. In:
Bio-technology applications for reforestation and biodiversity conservation Proceeding of the
8th International Workshop of BIO-REFOR, Kathamndu, Nepal, November 28-December 2,
1999. BIO-REFOR/IUFRO/SPDC, International Union of Forest Research Programme for
Development and Capacities, Austria, pp 256–260
40. Brondízio ES, Aumeeruddy-Thomas Y, Bates P, Carino J, Fernández-Llamazares Á, Ferrari
MF et al (2021) Locally based, regionally manifested, and globally relevant: indigenous and
local knowledge, values, and practices for nature. Annu Rev Environ Resour 46:481–509
41. Chaudhary RP (2000b) Biodiversity prospecting, systematics and intellectual property rights
(IPR). In: Jha PK, Karmacharya SB, Baral SR, Lacoul P (eds) Environment and agriculture: at
the crossroad of the new millennium. Ecological Society (ECOS), Nepal, pp 251–260
42. Cox PA, Balick MJ (1994) The ethnobotanical approach to drug discovery. Sci Am
270(6):82–87
43. Farnsworth NR (1988) Screening plants for new medicines. In: Wilson EO (ed) Biodiversity.
National Academy of Science, Washington, DC, pp 83–97
44. Oberthur S (2015) In: Oberthur S, Rosendal GK (eds) Global governance of genetic resources.
Routledge
45. Declaration M (1993) Mataatua declaration on cultural and intellectual property rights of indig-
enous peoples. Comm Hum Rights, Sub-Commission Prev Descrimination Prot Minor, Geneva
46. GoN (2015) Constitution of Nepal 2015. Singha Durbar, Kathmandu
47. Chaudhary RP, Uprety Y, Devkota S, Adhikari S, Rai SK, Joshi SP (2020) Plant biodiversity
in Nepal: status, conservation approaches, and legal instruments under new federal structure.
Plant Divers Nepal (March):167–206
48. Belbase N (1999) National implementation of the convention on biological diversity: policy
and legislative requirements. IUCN/Nepal
49. Warren DM (1996) Indigenous knowledge, biodiversity conservation, and development. In:
James VU (ed) Sustainable development in third world countries applied and theoritical per-
spectives, pp 81–88
50. Dowens D (1997) Using intellectual property as a tool to protect traditional knowledge: rec-
ommendations for next steps. Cent Int Environ Law:1
51. Uprety Y, Oli KP, Paudel KC, Pokharel DM, Thapa P, Chaudhary RP (2020) Accessing genetic
resources and sharing the benefits: the implications for research on biodiversity. In: Siwakoti
M, Jha PK, Rajbhandary S, Rai SK (eds) Plant diversity in Nepal. Botanical Society of Nepal,
Kathmandu, pp 2016–2224
Traditional Amchi Medicinal Practice in Trans-Himalaya of Nepal: Conservation… 327
52. Oli KP, Dhakal TD (2009) Access and benefit sharing from genetic resources and associated
traditional knowledge. International Centre for Integrated Mountain Development (ICIMOD),
Kathmandu
53. HMGN/MFSC (2002) Nepal biodiversity strategy. Ministry of Forest and Soil Conservation,
His Majesty’s Government of Nepal
54. GoN/MFSC (2014) Nepal national biodiversity strategy and action plan. Singha Durbar,
Kathmandu
55. MoFE (2018) Nepal’s sixth national report to the convention on biological diversity. Singha
Durbar, Kathamndu
56. Acharya R (1995) Biodiversity prospecting: prospects for private sector participation in the
Asia-Pacific region. Biodivers Conserv Asia Pasific Reg – Constraints Oppor Asian Dev Bank,
Manila, pp 367–388
57. ten Kate K, Laird SA (2000) The commercial use of biodiversity. Earthscan Publications
Ltd, London
58. Marshall E, Bagla P (1997) India applauds U.S. patent reversal. Science 277:1429
59. Gurung JD (1997) Gender dimensions in biodiversty management in Nepal. A Pap Present
a Natl Semin’ Conserving Bio-diversity Nepal’s Community for Organ by Dep for Nepal
Biodivers Action Plan 3
60. Chaudhary RP, Gupta VNP, Taylor RSL (2004) Cleyera japonica Thunb. var. wallichiana (DC.)
Sealy (Theaceae): a tea beverage plant of the Himalayas. Econ Bot 58(December):114–117
Website
1 Introduction
1.1 Medicinal Plants
Medicinal herbs have been used as a source of medicine to treat various ailments
from ancient times. Methodological approaches used in ethno-biological and ethno-
pharmacological surveys for collecting information on traditional applications of
plants and their natural products have improved over time. Plants with significant
medicinal significance that are utilized in primary health care have been the subject
of a number of research [1, 2]. Traditional medicine is used by 70–95% in Asia,
Africa, Latin America, and the Middle East for their fundamental health needs [3].
Natural products have emerged as an alternative form of treatment for a variety of
1.2 Desert Areas
Rangelands cover around 40% of the world’s geographical area and are just as vital
to the environment as rain forests. One-third of the world’s land surface is covered
by arid environments, is home to 14% of the world’s population, and produces a
major portion of global agriculture [10]. Rangelands are one of the most significant
ecosystems, offering pastoral and agro-pastoral inhabitants a wide range of services
and houses. They encompass a wide range of environments and biological commu-
nities in particular. They are also economically significant due to the abundance of
culinary, fodder, medicinal, and economic plant species. Rangeland plant biodiver-
sity is more visible and profound than in other habitats; therefore, any disturbance
has a greater impact [11]. Deserts, which have low and irregular rainfall, nutrient-
poor soils, and limited vegetation cover, cover about a third of the earth’s land sur-
face. Deserts offer a variety of features that can suit the needs of both local residents
and those in the nearby communities. Water, food, medicine, and raw resources are
among the advantages. Desert rangelands are subjected to harsh natural factors such
as high rates of soil erosion and extremely low rainfall patterns, which may be
induced in part by climate due to their geographic location [12].
The distribution, pattern, and quantity of plant species and communities in desert
environments have all been associated with physical ecological factors such as
water availability, soil chemistry, and anthropogenic disturbance [13]. Desert veg-
etation is adapted to extreme temperature and moisture changes, as well as edaphic
environments. Plants in this region grow slowly but respond fast to climatic changes,
and a major portion of the species can regrow even if there is not enough rain [14].
Traditional folk knowledge preservation, traditional medicine preference over oth-
ers, and biodiversity conservation have all gained popularity among many groups,
researchers, academicians, and policymakers. Traditional medical knowledge (a
source of low-cost herbal medicine) required effective techniques for its preserva-
tion in indigenous communities in order to ensure its long-term sustainability [15]
(Figs. 1 and 2).
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas 331
Fig. 1 Field pictorial photographs of desert dicot species: (a) Acacia jacquemontii, (b) Achyranthes
aspera, (c) Aerva javanica, (d) Amaranthus viridis, (e) Boerhavia diffusa, (f) Capparis decidua,
(g) Carthamus oxyacantha, (h) Chenopodium album, (i) Chenopodium murale, (j) Corchorus oli-
torius, (k) Cucumis melo var.agrestis, (l) Cuscuta reflexa, (m) Fagonia indica, (n) Heliotropium
europaeum, (o) Launaea procumbens
332 S. Majeed et al.
Fig. 2 Field pictorial photographs of desert dicot species: (a) Leptadenia pyrotechnica, (b) Malva
parviflora, (c) Peganum harmala, (d) Prosopis cineraria, (e) Prosopis juliflora, (f) Rhazya stricta,
(g) Rumex dentatus, (h) Salvadora oleoides, (i) Solanum surratense, (j) Tamarix aphylla, (k)
Tecomella undulate, (l) Trianthema portulacastrum, (m) Tribulus terrestris, (n) Withania coagu-
lans, (o) Ziziphus nummularia
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas 333
Ethnomedicinal appraisals of plant species are vital for conservation and preserva-
tion, as well as for the development of herbal medications. Plants have been used
extensively, and their therapeutic potential has been documented globally. Around
400–600 medicinal plants have been recognized as being used in Pakistan’s tradi-
tional health-care system. Pakistan is blessed with a diverse range of temperatures,
ecological zones, and topographical regions that are endowed with a variety of
medicinal plants, and the desert sections of Pakistan are believed to be as of yet
untouched in terms of ethnomedicinal surveys of medicinal plants [16]. The desert
supports a substantial human and livestock inhabitants. The majority of the popula-
tion is made up of nomads who follow the distribution of rainfall and the forage it
provides. The only source of income for the residents of the study region is livestock
keeping [17].
Despite the fact that 88% of Pakistan’s total land area is categorized as dry or
semi-arid, less focus is given to conducting research on desert environments [18].
Much research has been made in Pakistan on ethnomedicinal investigations in
northern hilly areas and plains, but the vegetation of the deserts has been ignored for
a number of reasons. Pakistan has a distinct geographical location in Asia’s Deserts.
Pakistan’s deserts have a peculiar plant richness, with an estimated 400–600 medic-
inal species. Based on a review of earlier studies, we uncovered only a few refer-
ences to medicinal plants from Pakistan deserts. Traditional plant-based treatments
are still more popular in desert communities than allopathic and biomedical medi-
cine. People with poor income and social status, in particular, must rely on tradi-
tional medication due to the unavailability of drugs and access to contemporary
health--care systems [19].
Sindh, but the majority of the studies have been conducted in Northern mountainous
regions of Pakistan, while the southern plain regions of Pakistan have either fewer
studies or are still unexplored. Among these areas, the deserts of Thal, Punjab, have
yet to be explored. Only a few studies from the Thal desert area have been docu-
mented in the literature [21, 22]. In addition, [23] reported 63 species from Sindh’s
northern deserts, while another study collected ethnomedicinal data on 51 plant
species from the Nara desert [17].
Although Thal desert area in Punjab province of Pakistan has a rich diversity of
medicinal plants and culturally important indigenous people that have a very strong
cultural link with medicinal plants used as herbal drugs, but the ethnobotanical doc-
umentation of important and precious medicinal plant species is still lacking behind.
The goal of this chapter is to deepen the knowledge of sustainable ethnomedicinal
uses of medicinal plants along with cultural recipes as practiced in the Thal desert.
An annotated list of dicot plants traditionally used as medicinal products has been
compiled along with a quantitative data analysis of specific herbal medicines used
in the desert area rangelands.
2 Methods
2.1 Data Documentation
Field surveys were conducted to acquire data from rural communities in Thal desert
zones in order to obtain quantitative data on dicot medicinal plants, followed by
participant observation and interviews. Traditional healers, herbalists, and local
residents are among the interviewers. Men and traditional healers provided the
majority of the medicinal data during open conversations. Because the majority of
the population speak Saraiki dialect, the interviews were conducted within this lan-
guage. A total of 87 people between the ages of 40 and 95 were interviewed, includ-
ing 74 men and 13 women. Plants’ local names, modes of use, parts used, and
medicinal uses were all recorded and represented in Table 2. Plants have been col-
lected, pressed, dried, poisoned, mounted, and submitted to the Herbarium of
Pakistan (ISL) for future research. Medicinal plant names were validated via the
plant list (www.theplantlist.org). Specimens were identified by Flora of Pakistan
and authenticated by comparing with herbarium specimens.
Table 2 Ethnomedicinal uses of dicot medicinal species inhabited in the deserts of Pakistan
Botanical name/ Common Flowering Part
S. No family name Habit season used Preparation Medicinal aspects FC*1 UR*2 RFC*3 CI*4 CV*5
1. Acacia Vachellia Tree July– Bark Paste The dried bark is 19 1 0.13 0.007 0.00007
jacquemontii jacquemontii September converted in the form of
Benth (Fabaceae) paste with water. The
paste is applied on the
cut by snake bite.
2. Achyranthes Prickly chaff Herb July– Root, Juice Juice of fresh leaves is 22 3 0.15 0.020 0.0005
aspera L. flower September leaves, used in aching teeth.
(Amaranthaceae) flowers Paste of spike is applied
as anti-venom against
scorpion sting. Root
paste is applied to
regulate the
menstruation cycle.
3. Aerva javanica Kapok bush Herb October– Whole Powder The flower is consumed 31 2 0.21 0.014 0.0003
(Burm.f) Juss.ex December plant against stomach
Schult. problems and stem is
(Amaranthaceae) applied for ear pain.
Inflorescence is used in
making cushions.
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas
11. Chenopodium Nettle-leaved Herb August– Aerial Decoction Decoction made from 35 1 0.24 0.007 0.00001
murale L. Goosefoot October parts the aerial part of plant
(Amaranthaceae) is used for
gastrointestinal
disorders.
(continued)
337
Table 2 (continued)
338
under inflammation.
(continued)
339
Table 2 (continued)
340
26. Mukia N/A Herb July– Leaves Powder Dry leaf powder is 34 1 0.23 0.007 0.0001
maderaspatana September mixed with butter and
(L.) M.Roem. used in dermatological
(Cucurbitaceae) problems.
27. Oxystelma Rosy Herb June– Whole Decoction A decoction of the plant 11 1 0.07 0.007 0.00004
esculentum (L.f) Milkweed September plant is useful as a gargle in
Sm. Vine infections of throat and
(Apocynaceae) mouth.
(continued)
341
Table 2 (continued)
342
affected by rheumatism.
The root bark is highly
effective in toothache
and gum inflammation.
Short cuttings of roots
and young shoots are
used as a Miswaak.
(continued)
343
Table 2 (continued)
344
43. Withania Poisonous Herb June– Roots Powder Roots are ground and 10 1 0.07 0.007 0.00004
somnifera (L.) gooseberry September mixed with honey to
Dunal prepare a tonic for hair
(Solanaceae) growth.
44. Ziziphus Wild jujube Shrub July– Leaves, Decoction The leaves are 14 3 0.10 0.020 0.0003
nummularia September bark antipyretic and reduce
(Burm.f.) Wight. obesity. Bark is used as
& Arn a remedy for diarrhea
(Rhamnaceae) and cures boils.
345
346 S. Majeed et al.
The ethnomedicinal data were confirmed from scientific literature catalogues such
as Pubmed, Web of Science, Google Scholar and Science Direct. The scientific
name of the desert species were authenticated with the comprehensive list of plant
species (https://wfoplantlist.org/).
2.3 Quantitative Analysis
Based on the number of informants, RFC is calculated to illustrate the local rele-
vance of indigenous species in the study area. It is calculated using the formula
provided by [24].
RFC = FC / N
Where NUR is the number of use citations in each category and Nt is the species
number taken as medicine.
This statistical index is calculated using the formula given by [27, 28].
CV NU / NC FC / N UR / N
NU is an abbreviation for the number of uses documented in the study. The word
NC denotes the total number of participants in the study. The relative frequency of
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas 347
citation is the second component, while the overall number of use reports is the
third. The numbers UR and N denote the total number of people participated.
CI = UR / N
UR signifies use reports, and N is the total number of informants. This index is used
to determine the number of uses, and the range of applications for each species.
3 Results
3.1 Informants Demography
Totally 87 informants were interviewed (Table 1). Dominant informants were males;
females were less in number because of the strict culture and traditions of the rural
areas of the study area. As compared to urban communities of the area, the rural
peoples in deserts have much knowledge about the herbal remedies. Mostly infor-
mants who were interviewed have average ages between 40 and 90 years (Table 1).
Even in previous studies regarding medicinal plants utilization in Pakistan, the same
fact was recorded regarding the male dominancy in informants [30–32]. Regarding
the informants’ ages, recent research has shown that elderly folks have more knowl-
edge than younger people [33]. In this study, most of the informants were illiterate
(46%), and it is also observed after interviews that the usage of medicinal plants was
dominant in desert areas as compared to urban areas as these results were similar in
accordance with the previously published literature [34, 35].
In the present study, 79% species are herbs. Other highly observed species are trees
12% and shrubs 8.6% (Fig. 3). Herbaceous plants are commonly used in previous
reports of [36]. Herbs are used most commonly in making herbal medicine due to
the wide distribution and presence of bioactive phytochemicals in them [37, 38].
The plant species used as medicine is mentioned in Table 3 with complete infor-
mation about the part used, mode of administration and their recipes. In this study,
44 medicinal plants classified to 22 families were recorded (Fig. 4). The most domi-
nant family was Amaranthaceae (six species) followed by Fabaceae and Solanaceae
Table 3 Informant consensus factor of plants and diseases treated in the desert area
Total number of Total number of plants that treat this
Disease categories disease disease ICF
Cardiovascular disorder 3 5 1.00
Digestive disorders 21 33 0.60
Eye disorders 3 4 0.50
Fever 3 4 0.50
Gastro intestinal 3 4 0.50
disorders
Gynecological disorders 7 8 0.17
Hormonal disturbance 2 3 0.25
Liver disorders 4 7 1.00
Musco-skeleton disorder 7 10 0.50
Oral disorders 7 11 0.67
Urinary tract disorders 10 15 0.56
Respiratory diseases 11 18 0.70
Sexual diseases 4 5 0.33
Skin disorders 18 35 1.00
Wounds 2 3 0.50
with five species each, Asteraceae, Cucurbitaceae, and Malvaceae (three species
each), Polygonaceae, and Apocynaceae, Boraginaceae, and Zygophyllaceae (two
species each) (Table 2). The family Amaranthaceae has a high potential for medici-
nal plants as previously reported by [39] as this family and its species are widely
distributed in the world desert areas [40]. Fabaceae, Solanaceae, and Asteraceae are
also effective in the treatment of common ailments [41, 42].
350 S. Majeed et al.
The ways medicinal plants prepared for utilization are mentioned in Table 1. The
plant parts used in this study are leaves, whole plant, and stem. Roots, seeds, and
fruits of some trees and shrubs are also used to make herbal medicine. The most
frequently used part of the medicinal plant is leaves (28%) followed by whole plant
(19%), stem (11%), fruits (10%), roots and flower (8% each), roots (7%), and bark
(6%) (Fig. 5). Leaves are the most used part of the plant in making herbal medicine
as most of the people belonging to rural communities of Pakistan utilize them [30,
36]. Also, the leaves contain a large amount of different phytochemicals and also
plays a vital role during photosynthesis process [43]. The same results were docu-
mented in studies carried out in the previous literature of [44]. The stem leaves and
flowers of different medicinal plants are used for treating several diseases such as
digestive disorders, hypertension, colic, respiratory disorders, and others [45].
In this study, herbal medicine is utilized in the form of decoction, powder, paste,
juice, infusion, poultice, extract, and raw form (Fig. 6). The majority of these medi-
cines are made in the form of decoction (25%) followed by powder (23%), paste
(20%), juice (18%), and raw form (14%). Decoction is considered to be the most
mode of administration in previous studies conducted by [46, 47]. It is easily made
by simply boiling plant parts in water. Powder form of plant is also mixed with but-
ter to treat skin problems (e.g., Mukia maderaspatana). Similar results were
obtained from previous studies reported by [48]. Traditional therapies for joint pain,
fever, muscular pain, cramps, wounds, and other ailments include oil, juice, infu-
sions, and paste made from various herbal remedies [49].
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas 351
3.6 Quantitative Analysis
The highest RFC value commonly refers to utilization, priority, and popularity of
plants listed by the informants for curing the specific ailment. Highest RFC values
are observed in Salvadora oleoides (0.53), Withania coagulans (0.52), and Fagonia
indica (0.50) (Table 2). The most cited species were most importantly used by peo-
ple for medicinal purposes [40]. The plants with the highest RFC value should be
involved in pharmacological, phytochemical, and biological actions [51]. Prosopis
352 S. Majeed et al.
3.6.3 Cultural Value
In this study, CV values ranges from 0.01 to 0.00001. Prosopis cineraria shows the
highest value CV = 0.010 followed by Salvadora oleoides CV = 0.0068 and Fagonia
indica with CV = 0.0044. The highest CV values indicate that these plants have
more than one medicinal use; for example, Prosopis cineraria is used in treating
leprosy, bronchitis, asthma, leukoderma, dysentery, hemorrhoids, and muscle
cramps (Table 2). The lowest CV would be seen in Chenopodium murale
(CV = 0.0001) followed by Amaranthus viridis, Carthamus oxyacantha, Peganum
harmala, Solanum surratense with the same CV = 0.0002 lowest value indicates
single uses of plant species.
3.7 Study Limitations
4 Conclusion
This chapter demonstrated the rich diversity of medicinal plants used to cure a wide
range of illnesses in desert settings. The chapter was prepared to document ethno-
medicinal knowledge in the desert rangeland using quantitative indices. Totally 44
medicinal species from 22 families have been reported to be utilized to treat a vari-
ety of disorders. The leaves were reported to be the most commonly utilized plant
part (28%) while herbs were the most commonly used life form (79%) and decoc-
tion was the most commonly used route of administration (25%). Due to the obvi-
ous growing awareness of medicinal plants and the relevance of ethnomedicinal
research, it is considered critical to record traditional medicinal plant knowledge
gathered from indigenous peoples of desert areas before it is lost forever. The
research establishes a baseline for future phytochemical and pharmacological
screening to evaluate the efficacy of medicinal plants in the development of herbal
drugs. It is thus recommended that policies for conservation of important medicinal
plant species should be planned.
Acknowledgement The authors would like to thank the Herbarium of Pakistan, Quaid-i-Azam
University, (ISL) for identification of plant specimens and providing lab facilities. We are also
grateful to Higher education commission of Pakistan for funding under project No NRPU-7837.
Conflict of Interest The authors declare no potential conflict of interest regarding publication of
this book chapter.
References
1. Kapoor L (2017) Handbook of ayurvedic medicinal plants: herbal reference library. Routledge,
Taylor and Francis group, CRC Press. Florida, United States of America
2. Iwu MM (2014) Handbook of African medicinal plants. CRC Press, Boca Raton
3. Van Wyk B-E, Wink M (2017) Medicinal plants of the world. CABI, Wallingford
4. Kewessa G, Abebe T, Demessie A (2015) Indigenous knowledge on the use and management
of medicinal trees and shrubs in Dale district, Sidama zone, Southern Ethiopia. Ethnobot Res
Appl 14:171–182
5. Tadesse A, Kagnew B, Kebede F (2018) Ethnobotanical study of medicinal plants used to treat
human ailment in Guduru District of Oromia Regional State. Ethiopia J Pharmacogn Phytother
10(3):64–75
354 S. Majeed et al.
6. Aziz MA, Khan AH, Adnan M, Izatullah I (2017) Traditional uses of medicinal plants reported
by the indigenous communities and local herbal practitioners of Bajaur Agency, Federally
Administrated Tribal Areas, Pakistan. J Ethnopharmacol 198:268–281
7. Abbas Z, Khan SM, Alam J, Khan SW, Abbasi AM (2017) Medicinal plants used by inhabit-
ants of the Shigar Valley, Baltistan region of Karakorum range-Pakistan. J Ethnobiol Ethnomed
13(1):53
8. Aziz MA, Khan AH, Adnan M, Ullah H (2018) Traditional uses of medicinal plants used
by indigenous communities for veterinary practices at Bajaur Agency, Pakistan. J Ethnobiol
Ethnomed 14(1):11
9. Shinwari ZK (2010) Medicinal plants research in Pakistan. J Med Plants Research 4(3):161–176
10. Gamoun M, Belgacem AO, Louhaichi M (2018) Diversity of desert rangelands of Tunisia.
Plant Divers 40(5):217–225
11. Gamoun M, Louhaichi M (2021) Botanical composition and species diversity of arid and des-
ert rangelands in Tataouine, Tunisia. Land 10(3):313
12. Bidak LM, Kamal SA, Halmy MWA, Heneidy SZ (2015) Goods and services provided by
native plants in desert ecosystems: examples from the northwestern coastal desert of Egypt.
Glob Ecol Conserv 3:433–447
13. Enright NJ, Miller BP, Akhter R (2005) Desert vegetation and vegetation-environment rela-
tionships in Kirthar National Park, Sindh, Pakistan. J. Arid Environ 61(3):397–418
14. Ansari KA, Mahar AR, Malik AR, Sirohi MH, Saand MA, Simair AA, Mirbahar AA (2017)
Impact of grazing on plant biodiversity of desert area of district Khairpur, Sindh, Pakistan. J
Anim Plant Sci 27(6):1931–1940
15. Yaseen G, Ahmad M, Potter D, Zafar M, Sultana S, Mir S (2018) Ethnobotany of medicinal
plants for livelihood and community health in deserts of Sindh-Pakistan. In: Plant and human
health, vol 1. Springer, Cham, pp 767–792
16. Yaseen G, Ahmad M, Sultana S, Alharrasi AS, Hussain J, Zafar M (2015) Ethnobotany of
medicinal plants in the Thar Desert (Sindh) of Pakistan. J Ethnopharmacol 163:43–59
17. Qureshi R, Bhatti GR (2008) Ethnobotany of plants used by the Thari people of Nara Desert,
Pakistan. Fitoterapia 79(6):468–473
18. Shaheen H, Qureshi R, Akram A, Gulfraz M (2014) Inventory of medicinal flora from Thal
desert, Punjab, Pakistan. Afr J Tradit Complement Altern Med 11(3):282–290
19. Yaseen G, Ahmad M, Shinwari S, Potter D, Zafar M, Zhang G, Sultana S (2019) Medicinal
plant diversity used for livelihood of public health in deserts and arid regions of Sindh-
Pakistan. Pak J Bot 2(31):2409–2419
20. Mahmood A, Malik RN, Shinwari ZK, Mahmood A (2011) Ethnobotanical survey of plants
from Neelum, Azad Jammu and Kashmir, Pakistan. Pak J Bot 43(105):10
21. Bhatti GR, Qureshi R, Shah M (2001) Ethnobotany of Qadanwari of Nara Desert. Pak J Bot
33:801–812
22. Panhwar AQ, Abro H (2007) Ethnobotanical studies of Mahal Kohistan (Khirthar national
park). Pak J Bot 39(7):2301–2315
23. Qureshi R, Bhatti GR, Memon RA (2010) Ethnomedicinal uses of herbs from northern part of
Nara desert, Pakistan. Pak J Bot 42(2):839–851
24. Fatima A, Ahmad M, Zafar M, Yaseen G, Khan MPZ, Butt MA, Sultana S (2017)
Ethnopharmacological relevance of medicinal plants used for the treatment of oral diseases in
Central Punjab-Pakistan. J Herb Med 12:88–110
25. Logan MH (1986) Informant consensus: a new approach for identifying potentially effec-
tive medicinal plants. In: Plants in indigenous medicine and diet: biobehavioral approaches.
Redgrave Publishing Company, Bedford Hills, p 91
26. Canales M, Hernández T, Caballero J, De Vivar AR, Avila G, Duran A, Lira R (2005) Informant
consensus factor and antibacterial activity of the medicinal plants used by the people of San
Rafael Coxcatlán, Puebla. Méx J Ethnopharmacol 97(3):429–439
27. Tardío J, Pardo-de-Santayana M (2008) Cultural importance indices: a comparative analysis
based on the useful wild plants of Southern Cantabria (Northern Spain). Econ Bot 62(1):24–39
Appraisal of Medicinal Plants Diversity Inhabited in Deserts Areas 355
28. Zhang Y, Xu H, Chen H, Wang F, Huai H (2014) Diversity of wetland plants used traditionally
in China: a literature review. J Ethnobiol Ethnomed 10(1):72
29. Menendez-Baceta G, Aceituno-Mata L, Reyes-García V, Tardío J, Salpeteur M, Pardo-de-
Santayana M (2015) The importance of cultural factors in the distribution of medicinal plant
knowledge: a case study in four Basque regions. J Ethnopharmacol 161:116–127
30. Malik K, Ahmad M, Zhang G, Rashid N, Zafar M, Sultana S, Shah SN (2018) Traditional plant
based medicines used to treat musculoskeletal disorders in Northern Pakistan. Eur J Integr
Med 19:17–64
31. Butt MA, Ahmad M, Fatima A, Sultana S, Zafar M, Yaseen G, Ashrfa MA, Shinwari ZK,
Kayani S (2015) Ethnomedicinal uses of plants for the treatment of snake and scorpion bite in
Northern Pakistan. J Ethnopharmacol 168:164–181
32. Ahmad M, Sultana S, Fazl-i-Hadi S, Ben Hadda T, Rashid S, Zafar M, Khan MA, Khan PZ,
Yaaseen G (2014) An ethnobotanical study of medicinal plants in high mountainous region of
Chail valley (District Swat-Pakistan). J Ethnobiol Ethnomed 10(1):36
33. Kayani S, Ahmad M, Sultana S, Shinwari ZK, Zafar M, Yaseen G, Hussian M, Bibi T (2015)
Ethnobotany of medicinal plants among the communities of Alpine and Sub-alpine regions of
Pakistan. J Ethnopharmacol 164:186–202
34. Bibi T, Ahmad M, Tareen RB, Tareen NM, Jabeen R, Rehman S-U, Sultan S, Zafar M, Yaseen
G (2014) Ethnobotany of medicinal plants in district Mastung of Balochistan province-
Pakistan. J Ethnopharmacol 157:79–89
35. Bibi S, Sultana J, Sultana H, Malik RN (2014) Ethnobotanical uses of medicinal plants in the
highlands of Soan Valley, Salt Range, Pakistan. J Ethnopharmacol 155(1):352–361
36. Ullah M, Khan MU, Mahmood A, Malik RN, Hussain M, Wazir SM, Daud M, Shinwari
ZK (2013) An ethnobotanical survey of indigenous medicinal plants in Wana district South
Waziristan agency, Pakistan. J Ethnopharmacol 150(3):918–924
37. Uniyal SK, Singh K, Jamwal P, Lal B (2006) Traditional use of medicinal plants among the
tribal communities of Chhota Bhangal, Western Himalaya. J Ethnobiol Ethnomed 2(1):14
38. Sanz-Biset J, Campos-de-la-Cruz J, Epiquién-Rivera MA, Canigueral S (2009) A first sur-
vey on the medicinal plants of the Chazuta valley (Peruvian Amazon). J Ethnopharmacol
122(2):333–362
39. Santhiya E, Banu AR, Anushiya DC, Vengadeswari A, Mahesh R (2021) Survey of medicinal
plants in Kariyamanikapuram, Nagercoil, Kanyakumari District, Tamil Nadu. India Bot Rep
10(2):4–9
40. Koti M, Katrahalli K (2021) Wild edible fruits and vegetables of Yadahalli Chinkara Wildlife
Sanctuary, Bagalkot, Karnataka, India. J Glob Biosci 10(9):8998–9008
41. Menendez-Baceta G, Aceituno-Mata L, Molina M, Reyes-García V, Tardío J, Pardo-de-
Santayana M (2014) Medicinal plants traditionally used in the northwest of the Basque
Country (Biscay and Alava), Iberian Peninsula. J Ethnopharmacol 152(1):113–134
42. Akerreta S (2009) Etnobotánica farmacéutica en Navarra: del uso tradicional de las plantas
medicinales a su evidencia científica. Faculty of Science. 831: University of Navarra Pamplona
43. Ahmad M, Khan MPZ, Mukhtar A, Zafar M, Sultana S, Jahan S (2016) Ethnopharmacological
survey on medicinal plants used in herbal drinks among the traditional communities of
Pakistan. J Ethnopharmacol 184:154–186
44. Shil S, Choudhury M, Das S (2014) Indigenous knowledge of medicinal plants used by the
Reang tribe of Tripura state of India. J Ethnopharmacol 152(1):135–141
45. Leporatti M, Ivancheva S (2003) Preliminary comparative analysis of medicinal plants used in
the traditional medicine of Bulgaria and Italy. J Ethnopharmacol 87(2–3):123–142
46. Bano A, Ahmad M, Hadda TB, Saboor A, Sultana S, Zafar M, Khan MPZ, Arshad M, Ashraf
MA (2014) Quantitative ethnomedicinal study of plants used in the skardu valley at high alti-
tude of Karakoram-Himalayan range, Pakistan. J Ethnobiol Ethnomed 10(1):43
47. Özcan MM, Ünver A, Uçar T, Arslan D (2008) Mineral content of some herbs and herbal teas
by infusion and decoction. Food Chem 106(3):1120–1127
356 S. Majeed et al.
48. Okwu DE, Josiah C (2006) Evaluation of the chemical composition of two Nigerian medicinal
plants. Afr J Biotechnol 5(4):357–361
49. Schempp C, Winghofer B, Lüdtke R, Simon-Haarhaus B, Schöpf E, Simon J (2000) Topical
application of St John’s wort (Hypericum perforatum L.) and of its metabolite hyperforin
inhibits the allostimulatory capacity of epidermal cells. Br J Dermatol 142(5):979–984
50. Kartal M (2007) Intellectual property protection in the natural product drug discovery, tradi-
tional herbal medicine and herbal medicinal products. Phytother Res 21(2):113–119
51. Mukherjee PK, Nema NK, Venkatesh P, Debnath PK (2012) Changing scenario for promotion
and development of Ayurveda–way forward. J Ethnopharmacol 143(2):424–434
Bush Medicinal Plants of the Australian
Wet Tropics and Their Biodiscovery
Potential
1 Introduction
The “Wet Tropics” encompasses 450 km along the northeast coast of Australia,
covering approximately 8940 km2 of Australian tropical rainforest with latitude
−17° 29′ 59.99″ South and longitude 145° 44′ 59.99″ East (Fig. 1) [1]. The Wet
Tropics stretch from low sea level to high mountains, such as Mt. Bartle Frere,
which is at an altitude of 1622 m above sea level. It is listed as one of the 35 inter-
national global hotspots in 2010 [2], and it covers 0.12% of Australia [3]. The Wet
Tropics bioregion on large scale has seven different vegetation groups: rainforest,
sclerophyll and sclerophyll rainforest transition vegetation groups, sclerophyll for-
ests and woodlands, vegetation complex and mosaics, shrublands and heathlands,
non-woody vegetation, and mangroves with 24 plant communities [4, 5]. The Wet
Tropics experience an early wet season from October to December and a monsoon
season from January to April [6]. The Wet Tropics bioregion is home to more than
2800 plant species, out of which more than 700 species (i.e., approximately 25%)
are endemic to the area [2]. The “Wet Tropics” is significant not only from an eco-
logical point of view but also for preserving important historical, cultural, and
anthropogenic activities, including Aboriginal people’s vast knowledge about edi-
ble and medicinal plants [7].
At present, about 20 Aboriginal communities comprising 120 clans speaking
eight languages live in the “Wet Tropics” [7]. These Rainforest Aboriginal people
have lived in harmony with nature for thousands of years and their knowledge in
utilising plants for food, shelter, and medicine is commendable. They are the pio-
neers in managing biodiversity and landscapes, through activities such as plant
Fig. 1 Map of Australia showing the Wet Tropics World Heritage Area
cultivation and active traditional mosaic burning, even before the onset of Australia’s
early colonial period (1788–1850) [8–10]. For instance, cultivation of cycads and
fruit trees and mosaic burning among Yalanji country people in the northern Wet
Tropics is well-documented [9], and their cultural burning is valued and incorpo-
rated into the forest management practices. At least two seats in the Wet Tropics
Management Board of Directors are reserved for Aboriginal people as custodians
and traditional knowledge owners [7]. About 87.5% of the Wet Tropics World
Heritage Area is constituted by native titles and other rainforest Aboriginal land
interests [11]. Two native title representative bodies (North Queensland Land
Council and Cape York Land Council) in the Wet Tropics region are responsible for
protecting and securing native titles [12].
Aboriginal peoples’ ecological legacy is recognized with increasing administra-
tive support for the documentation, preservation, and promotion of their ancestral
knowledge, through establishing rangers’ groups, traditional owner leadership
groups, indigenous protected areas, and research collaborations with researchers,
scientists, and various academic institutions across Australia. Tropical Indigenous
Ethnobotany Centre (TIEC) is one such example of indigenous initiatives. TIEC is
the first indigenous-driven ethnobotanical research center in Australia, established
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 359
Fig. 2 An Indigenous Ethnobotanist (Mr. Gerry Turpin at work) collecting herbarium specimens.
He is leading a Tropical Indigenous Ethnobotany Centre (TIEC) at Australian Tropical Herbarium
(ATH), James Cook University, Cairns. (Courtesy: Phurpa Wangchuk)
360 K. Yeshi and P. Wangchuk
troops. More than 500 tonnes of dried and powdered leaves were exported to
German and Swiss pharmaceutical industries in 1989 [15].
While the Rainforest Aboriginal communities still practice bush food and bush
medicine cultures, only a limited number of plants have been explored for commer-
cial activities. Some Traditional Knowledge requires rediscovery, and TIEC is
working closely with various Aboriginal communities to document and protect their
bush medicine knowledge. Part of this documentation identifies plants on their lands
that offer new business potential in the food, beverage, ornamental, health-
promoting, and nursery industries. There is an urgent need to develop a complete
inventory list of plants that grow in the Wet Tropics and determine their economic
values and market potential.
This chapter highlights 30 medicinal plants that grow in the ‘Wet Tropics’ region.
We identified only three medicinal plant species endemic to the Wet Tropics and
Cape York region. Many medicinal plants listed here are reported to grow in other
parts of Australia. The traditional uses, phytochemical content, and pharmacologi-
cal properties were extracted from the published documents, including journal arti-
cles, books, book chapters, and official websites. The botanical names of the plants
were confirmed using ‘The World Flora Online’ (https://wfoplantlist.org/plant-list).
The information on the distribution of plants was obtained using ‘The Australian
Virtual Herbarium’ (https://avh.chah.org.au/) and the ‘Discover Life’ (https://www.
discoverlife.org/20/q). The information gathered from these sources is presented
below individually.
2.1.1
Alphitonia excelsa (Fenzl) Reissek ex Benth. (Rhamnaceae)
Alphitonia excelsa is a medium-size tree with grey to golden-brown hairy buds and
stems, when young. It grows up to a height of 21 m. It is commonly called the ‘red
ash’ or ‘soapbush’ (because the wet leaves create a lather) or ‘leatherjacket.’
Ethnomedicine: People bath in warm infusions prepared from leaves to relieve
headache [16]. Leaves are also applied to eyesore [17]. While infusion prepared
from roots, bark, and wood is applied to the body to heal pain [17]. Wood and bark
decoction is effective for toothache [16]. Young shoot tips are chewed to ease stom-
ach upset [16]. Aboriginal people of Yaegl country use leaves to treat sores, wounds,
and skin infections [18].
Phytochemistry and Pharmacology: γ-sitoserol, nonanal, n-tetracontane, doco-
sane, 1,54-dibromotetrapentacontane, tetradecane, and hexadecane were identified
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 361
from leaves [18]. Betulinic acid was also isolated from its twigs [19]. The crude leaf
extract (dichloromethane and hexane extract) showed moderate antibacterial activ-
ity (Staphylococcus aureus) at MIC of 312.5–625 μg/mL and 1250–2500 μg/mL,
respectively [18]. Betulinic acid showed antiproliferative activity against human
ovarian cancer cell line A2780 with an IC50 value of 20.6 μM [19].
2.1.2
Clerodendrum floribundum R.Br. (Lamiaceae) (Fig. 3a)
Clerodendrum floribundum is a tall shrub or a small tree that produces black seed
surrounded by red calyxes, leading to the common name ‘lolly bush.’
Ethnomedicine: Decoction prepared from wood is used to treat aches and pains
[16]. A water extract made from the leaves was used on the skin to reduce itch-
ing [20].
Phytochemistry and Pharmacology: No phytochemical and pharmacological
studies reported for this plant.
2.1.3
Cymbopogon bombycinus (R.Br) Domain (Poaceae)
Fig. 3 Selected bush medicinal plant photos. (Courtesy: Paul Williams and Phurpa Wangchuk).
(a) Clerodendrum floribundum. (b) Excoecaria parvifolia. (c) Canarium australianum. (d)
Erythrophleum chlorostachys. (e) Flagellaria indica. (f) Macaranga tanarius
362 K. Yeshi and P. Wangchuk
(Mal), Jorroy (Mang), Guruguruny, and Gububu (Yarr) [21]. This grass is native to
Northern Territory.
Ethnomedicine: A whole grass is burnt and rubbed on the body for pain and
colds. In some cases, the grass is soaked in water and the liquid treats sore eyes [17].
Phytochemistry and Pharmacology: There is no record of phytochemical studies.
2.1.4
Ehretia saligna R.Br. (Boraginaceae)
Ehretia saligna is a shrub or a small tree that grows up to 6 m tall. It has common
names such as ‘Ehretia,’ ‘coonta,’ ‘peachwood,’ ‘peach bush,’ ‘false cedar,’ and
‘native willow.’ Aboriginal people call it Warlagarri [21]. This plant occurs in the
Gulf of Carpentaria and the coastal area of Northern Queensland.
Ethnomedicine: The latex and chopped inner bark are boiled, and the resulting
red liquid is applied to cuts and sores [22]. The decoction prepared from the wood
has been taken orally to alleviate aches and pains [16].
Phytochemistry and Pharmacology: No phytochemical and pharmacological
studies reported for this plant.
2.1.5
Excoecaria parvifolia Müll.Arg. (Euphorbiaceae) (Fig. 3b)
2.1.6
Pandanus spiralis R.Br. (Pandanaceae)
Ethnomedicine: The inner core of the upper section of the trunk of young trees is
eaten by Aboriginal people to heal stomach pain and diarrhea [24].
Phytochemistry and Pharmacology: Not studied for its phytochemical and phar-
macological activities.
2.1.7
Pteridium esculentum (G. Frost.) Cockayne (Dennstaedtiaceae)
2.1.8
Rhaphidophora australasica F.M.Bailey (Araceae)
2.1.9
Zieria smithii Jacks. (Rutaceae)
Zieria smithii is a robust shrub that grows up to 2 m high with glandular stems and
leaves, which emit a pungent smell when crushed. There are many common names
including ‘turmeric,’ ‘sandfly bush,’ ‘lanoline bush,’ ‘stunk wood,’ and ‘sandfly
zieria.’
Ethnomedicine: Leaves are crushed and then inhaled to cure headache, but
excessive use may make headache worse [28].
Phytochemistry and Pharmacology: Identified α-pinene, thymol, chrysanthe-
none, citronellol, safrole, bicyclogermacrene, and butylated hydroxytoluene from
its essential oil [29]. The hexane extract of Zieria smithii showed anti-bacterial and
anti-fungal activities [29].
364 K. Yeshi and P. Wangchuk
2.2.1
Calamus caryotoides A.Cum. Ex Mart. (Arecaceae)
2.2.2
Melaleuca quinquenervia (Cav.) S.T.Black (Myrtaceae)
2.2.3
Tephrosia varians (Bailey) C.T.White (Fabaceae)
2.3.1
Canarium australianum F.Muell. (Burseraceae) (Fig. 3c)
2.3.2
Cymbidium madidum Lindl. (Orchidaceae)
2.4.1
Ajuga australis R.Br. (Lamiaceae)
Ajuga australis is an herb, and it is also known by the common name ‘Australian
bugle,’ which is widespread over East Australian states.
Ethnomedicine: The whole bruised plant is mixed with hot water to make an
infusion and used for bathing sores and boils [30].
Phytochemistry and Pharmacology: Ajugapitin and (15R)-14,15-dihydro-15-
hydroxyajugapitin are isolated from aerial parts [38]. A crude foliar extract showed
contact toxicity against the crop pest Tetranychus urticae [39].
2.4.2
Brachychiton diversifolius R.Br. (Malvaceae)
Brachychiton diversifolius is a tree that grows to 3–25 m tall. The plant is com-
monly known as ‘Brachychiton,’ ‘kurrajong,’ and ‘northern kurrajong.’ In
Aboriginal languages, it is referred to as Birdba (Ngal, Nung), Dagdag (Jam),
Ngaliwurru, Nungali, Pinkyekper (Mal), Pinyengertpe (Mat), Dirlay (Mang,
Yang), Dakdakkin and Gulguldum in Warray [18]. It is often harvested for its
edible seeds and fibre. This tree is also spotted in Northern Queensland, Northern
Territories, and Western Australia. It usually grows along stony hills, riversides,
red sandy soils, basalt, and limestone areas. Seeds are usually cooked and eaten
but have low protein and fats [40, 41]. The plant is also harvested to sell as a
decorative plant [42].
Ethnomedicine: An extract prepared from the inner bark is used as an eye-
wash. Leaves, usually from the young plant, are pounded and soaked in water
for one hour, and the resultant infusion is used as body wash and a little liquid
placed in each ear. Such liquid is also used for treating skin lesions. A flexible
bark, stripped from the tree, serves as a handy bandage to wrap cuts and
wounds [28].
Phytochemistry and Pharmacology: The phytochemical analysis of the crude
ethanol extract of wood branches showed the presence of tannins, flavonoids,
alkaloids, saponins, phenols, and steroids [43]. The seed oil contains triacylg-
lycerols, such as linoleic and oleic acids, and amino acids [41]. Essential oils
from wood, bark, and leaves were analysed using gas chromatography and iden-
tified 21 methyl esters of fatty acid, out of which myristic acid in the wood and
palmitic acid in bark and leaves were major methyl esters of fatty acid [44].
There is no record of isolation of pure compounds from any plant parts of this
plant. The fatty acid fraction obtained from the wood, bark, and leaves showed
mild to moderate anti-bacterial (Bacillus subtilis and Sarcina lutea), anti-fungal
(Penicillium selerotigenum, Paecilomyces variotii, and Aspergillus niger), and
antioxidant activities [43, 44].
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 367
2.4.3
Erythrophleum chlorostachys (F.Muell.) Baill. (Fabaceae) (Fig. 3d)
2.4.4
Persoonia falcata R.Br. (Proteaceae)
Persoonia falcata is a shrub commonly called ‘Persoonia,’ ‘wild Pear,’ and ‘milky
Plum.’ In Aboriginal languages, it is called by different names as Warlwaya (Jam,
Ngal, Nung), Gilayi, Ngerrinytya, Ningggij (Mang), Gayabam (Yang), Warlaya,
Garttan, and Makbuny [21]. The plant is native to Northern Territory but also occurs
in Queensland and Western Australia.
Ethnomedicine: Fruits are eaten raw. Leaves and bark are considered medicinal.
An infusion of the leaves and barks are used as a treatment against sore throat and
colds [27]. An infusion from wood is applied externally to eyesores [16].
Phytochemistry and Pharmacology: The crude extract from the leaves and stem
showed anti-inflammatory activities by inhibiting pro-inflammatory cytokines in
the human peripheral blood mononuclear cells assay [47]. There is no report on its
phytochemicals.
2.5 Skin Disorders
2.5.1
Acacia falcata Willd. (Leguminosae)
Acacia falcata is a shrub or tree that grows about 2–5 m tall (erect or spreading).
The plant is commonly called a ‘sickle wattle.’ It also occurs as far as inland to the
tablelands in Queensland and on the central coast of New South Wales, including
Narooma. It usually grows in sclerophyll forests, particularly coastal areas [21].
Ethnomedicine: The tree’s bark is used by Aboriginal communities (Yaegl peo-
ple of North NSW) to prepare embrocation to treat cutaneous diseases [48, 49].
368 K. Yeshi and P. Wangchuk
Moreover, in the counties of Cumberland and Camden, the people used bark (based
on tannin content) to stupefy fish.
Phytochemistry and Pharmacology: Leaves contain alkaloids, phenolics, flavo-
noids, terpenoids, anthraquinones, and condensed tannins [48]. Exudate contains
glucuronic acid, galactose, arabinose, and traces of rhamnose [50].
A crude extract of leaves showed anti-bacterial activity against multi-drug-
resistant Staphylococcus aureus with the MIC value ranging from 250 to 1000ug/
mL. It is also anti-oxidative through DPPH (2,2-diphenyl-1-picryl-hydrazyl-
hydrate), ABTS (3-ethylbenzothiazoline-6-sulfonic acid), and FRAP (ferric reduc-
ing antioxidant power) assays [48]. There is no record of isolation of pure compounds
from any parts of this plant.
2.6.1
Ageratum conyzoides (L.) L. (Compositae)
Ageratum conyzoides is an erect branched and hairy annual herb, that grows up to
30–60 cm tall. Herb is commonly called ‘billygoat weed’ or ‘billygoat plant.’ It is
not native to Australia, where it is a weed, but it originates from Central and South
America.
Ethnomedicine: Aboriginal people of Northern Queensland mash the whole
plant and then apply it to wounds to promote healing [51].
Phytochemistry and Pharmacology: 5′-methoxy nobiletin, coumarin, and eupal-
estin are isolated from aerial parts [52]. All these three compounds exhibited anti-
inflammatory activity in a carrageenan-induced inflammation of mouse model by
inhibiting inflammatory cytokines such as IL-6, IFN-γ, p65 NF-kB, and MAPK
activation. Coumarin also increases the level of anti-inflammatory cytokine IL-10
[52]. The activity shown by these compounds is relatable to the wound-healing
property of the herb. Other active constituents: 5,6,7,8,3′,4′,5′-heptamethoxyflavone
[53], coumarin, [54, 55], lycopsamine, dihydrolycopsamine, and acetyl-lycopsamine,
stigmasterol, precocene II, encecalol, demethoxyencecalol, sesamin, linderoflavone
B, 3′-hydroxy-5,6,7,8,4′,5′-hexamethoxyflavone, 2-hydroxycinnamic acid, and
3,4-dihydrocoumarin [56]; eupalestin, 5,6,7,5′-tetramethoxy-3′,4′-
methylenedioxyflavone, 5,6,7,3′,4′,5′-hexamethoxyflavone, ageconyflavone C [57];
ageconyflavones A-B, sinensetin, and 5,6,7,8,3′,5′-hexamethoxy-4′-
hydroxyflavone [58].
2.6.2
Cynanchum viminale subsp. australe (P.I.Forst.) Liede &
Meve (Apocynaceae)
Ethnomedicine: Aboriginal people use latex of this plant to treat bleeding wounds
sores, skin lesions, and cuts [59].
Phytochemistry and Pharmacology: There is no record of any phytochemical and
pharmacological studies on this plant so far.
2.6.3
Dodonaea polyandra Merr. & L.M.Perry (Sapindaceae)
Flagellaria indica is a semi-woody climbing plant with stems 10–15 m long and
1–2 cm thick. It is commonly called as ‘false rattan.’
Ethnomedicine: Astringent leaves are used for treating wounds [63].
Phytochemistry and Pharmacology: There is no record of analysing phytochemi-
cal constituents of this plant, but the aqueous leaf extract had phenolic content of
65.88 ± 1.84 mg gallic acid equivalent/g. The crude extract showed a protective
effect against carbon tetrachloride (CCl4)-mediated liver injury by suppressing oxi-
dative stress markers (4-hydroxynonenal and 8-hydroxydeoxyguanosine) and pro-
inflammatory cytokines, such as TNF-α, IL-6, and PGE2 [64].
2.6.4
Grevillea coriacea McGill (Proteaceae)
2.6.5
Grevillea striata R.Br. (Proteaceae)
Grevillea striata is a 10 m tall tree with dark, persistent fissured bark. It is com-
monly known as ‘Grevillea’ and ‘beefwood.’ In Aboriginal languages, it is known
as Iyltenty, Lyent, Gulburru, Jirrirndi, Jawilyi, Piltentye, Wubgaji, Yiltilypa, Iltilpa,
Yiltilypa and Yilykinji, and ltyantye [21]. This species is easily distinguishable from
other species of the same genus as it has long strap-like leaves, cream flowers, and
a unique growth form. This plant is native to Northern Territory. It also occurs near
the coast in Queensland and the Northwest of Western Australia [21].
Ethnomedicine: Fruit is edible. Sap and wood are considered medicinal. The sap
is used to treat sores and wounds, and charcoal made from wood is used for stop-
ping bleeding from certain spear wounds [17].
Phytochemistry and Pharmacology: Striatol is isolated from heartwood [66].
Striatol showed a potent inhibition of PM Ca2+-ATPase, with an IC50 value of
16 μM [67].
2.6.6
Macaranga tanarius (L.) Mull.Arg. (Euphorbiaceae) (Fig. 3f)
Macaranga tanarius is a deciduous shrub or small tree and grows up to 4–15 m tall.
It is commonly called ‘macaranga.’ It is also known as Dokoo-ral among the
Aboriginal people of Yirrganydji and Djabugay.
Ethnomedicine: Aboriginal people apply red sap to cure skin wounds and deep
cuts [28]. Aboriginal people also use leaves for cooking and wrapping food.
Phytochemistry and Pharmacology: Diterpene macarangonol isolated from the
leaf and stem [68]. Macaflavanones A-G, nymphaeol, nymphaeol C, tanariflavanone
A & B, and kolavenol are isolated from leaves [69, 70]. Macaflavanone G showed
toxicity against the KB cells (epidermal nasopharyngeal carcinoma, human cell
line) and A549 cell lines (lung cancer cell line) using 3-[4,5-dimethylthiazol-2
-yl]-2,5- diphenyltetrazolium bromide (MTT) assay with IC50 values of 12.3 and
13.4 (3.0 and 2.1 μM for KB and A549 cells), respectively [69, 70].
2.6.7
Melaleuca leucadendra L. (Myrtaceae)
Melaleuca leucadendra is a large tree often called paperback tree and prefers to
grow in the moist forest where fresh water is available all year round. Leaves
become hairless upon maturity, which is why sometimes it is also called ‘white-
leafed tea tree.’
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 371
2.6.8
Pterocaulon serrulatum (Montrouz.) Guillaumin (Compositae)
Pterocaulon serrulatum is a strongly scented, hairy perennial herb that grows about
40–90 cm tall. It is commonly known as ‘ragweed’ or ‘pintye-pintye.’ In Aboriginal
languages, it is called Tjungarai, Alworm-angka-ina, and Penja-pemja [77].
Ethnomedicine: Aboriginal people rub the whole plant over wounds and stuff
them with this herb [17].
Phytochemistry and Pharmacology: Ayapin, 5-methoxy-6,7-
methylenedioxycoumarin, 6,7,8-trimethoxycoumarin, 5,7-dihydroxyflavanone, and
5-methoxyscopoletin were isolated from aerial parts [78]. Pharmacological data is
not reported for this species.
2.6.9
Sterculia quadrifida R.Br. (Malvaceae)
2.7 Miscellaneous
2.7.1
Doryphora aromatica (F.M.Bailey) L.S.Sm. (Atherospermataceae)
About two-thirds of medicinal plant species used worldwide are sourced from their
natural habitats [83]. Australia’s tropical forests, including ‘Wet Tropics,’ are home
to vast numbers of medicinal plant species. Many plants growing in the Wet Tropics
are affected by climate change and to survive, and the plants produce interesting and
higher concentration of biomolecules that could be exploited for pharmaceutical
research and development [84]. Most of these medicinal species remain least
explored for their phytopharmaceutical properties and have huge potential for dis-
covering natural product solutions for infectious and chronic diseases [85]. For
instance, only 16 out of 30 plant species included in this chapter have been studied
for their phytochemical or pharmacological properties. Rest 14 medicinal plants are
either not studied or only partially studied. Of the studied plants, 71 compounds
have been isolated from 16 medicinal plants included in this chapter. Out of 71
compounds, 10 compounds showed potent pharmacological activities, including
anti-inflammatory, anti-microbial, antioxidant, and anti-proliferative activities (see
Fig. 4 for the representative structures of bioactive molecules isolated from Wet
Tropics medicinal plants). Many compounds remain unassessed for their pharmaco-
logical properties.
Compounds such as coumarin from Ageratum conyzoides, ellagitannin and casu-
arinin from Melaleuca leucadendron, grandinin from Melaleuca quinquenervia,
and hexadecenoic acid from Sterculia quadrifida showed potent anti-inflammatory
activities [32, 52, 72, 80]. These medicinal plants are used by Aboriginal people to
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 373
Diosphenol Casuarinin
Eupalestin
Fig. 4 Chemical structures of selected bioactive compounds isolated from Wet Tropical medici-
nal plants
heal wounds and inflammations, which makes it interesting. Thus, these compounds
are worth assessing further to develop wound-healing drugs or creams.
Macaflavanone G isolated from the leaves of Macaranga tanarius and diosphenol
from Excoecaria parvifolia (Fig. 4) are found to be toxic against human cancer cell
lines [23, 69, 70]. Betulinic acid isolated from leaves and twigs of Alphitonia excelsa
(Fig. 4) also showed anti-proliferative activity against human ovarian cancer cell
lines [19]. These compounds could be potential candidates for developing anti-
cancer drugs. The crude extract obtained from the leaves of Acacia falcata showed
potent anti-bacterial activity, including against multi-drug resistant species
Staphylococcus aureus, and thus, this medicinal plant may contain promising anti-
microbial drug leads.
Overall, many bush-medicinal plants included in this chapter remain less
explored for their phytochemical profiles and pharmaceutical applications. This
could be partially due to limited accessibility as their collection is strictly regulated
by The Convention on Biological Diversity [86], Queensland Biodiversity Act and
other local state and federal Biodiversity Protection Acts, requiring collection per-
mits for accessing and studying the plants.
Recently, there has been a renewed interest among TOs aspiring to collaborate
with scientists to investigate their bush food and medicine and add value to it. A few
374 K. Yeshi and P. Wangchuk
4 Conclusion
Wet Tropics has both cultural and ecological significance to Australia. Culturally,
“Wet Tropics” is home to many indigenous communities, whose ancestors were
highly skilled and knowledgeable explorers of the Wet Tropics vegetation. They
learned to survive for millennia utilising their rich flora and fauna in food, medicine,
shelter, and protection. Even today, the Aboriginal communities possess vast tradi-
tional knowledge of plants, and they play a critical role in conserving the Rainforests
of the Wet Tropics of Australia. Most medicinal plants growing in the “Wet Tropics”
are either native or endemic to Australia, and they are least explored for their phy-
tochemical properties. Of many tropical medicinal plants of Australia, 30 medicinal
plants included in this chapter grow in the Wet Tropics region. Of 71 compounds
that have been isolated from 16 of these medicinal plants, ten compounds showed
potent pharmacological activities, including anti-inflammatory, anti-microbial, anti-
oxidant, and anti-proliferative. Thus, “Wet Tropics” presents an excellent resource
for drug discovery. Traditional custodians and their younger generations have
started documenting their traditional knowledge and establishing research collabo-
ration with bigger academic institutions across Australia, especially with James
Cook University. Such initiatives are a welcoming sight as they could validate the
traditional uses of wild plants, develop bush foods and health-promoting products,
and provide novel drug lead molecules for treating many diseases.
Acknowledgement We would like to thank Mr. Gerry Turpin (Ethnobotanist) of TIEC, JCU and
Paul Williams (Botanist) of Vegetation Management Science, Malanda, Queensland for their sug-
gestions and comments.
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 375
References
1. WHC (n.d.) World heritage conservation. Wet Tropics of Queensland. Retrieved on February
28, 2022 from https://whc.unesco.org/en/list/486/
2. Wet Tropics Nanagement Authority. Endemic and rare species. Retrieved on February 12,
2022 from https://www.wettropics.gov.au/endemic-species
3. Wet Tropics Management Authority (2016) State of wet tropics report 2015–2016. Ancient,
endemic, rare and threatened vertebrates of the wet tropics. https://www.wettropics.gov.au/
site/user-assets/docs/sowt2015-16b5-lres.pdf
4. Webb LJ, Tracey JG (1994) The rainforests of northern Australia. In: Groves RH (ed) Australian
vegetation, 2nd edn. Cambridge University Press, pp 87–130
5. Sanderson R (2008) Re-writing the history of Australian tropical rainforests: ‘alien invasives’
or ‘ancient indigenes’? Environ Hist 14:165–185
6. CSIRO (2019) Bureau of Meteorology and the CSIRO. Regional weather and climate guides.
Retrieved on February 28, 2022 from http://www.bom.gov.au/climate/climate-guides/
guides/027-Wet-Tropics-QLD-Climate-Guide.pdf
7. Roberts P, Buhrich A, Caetano-Andrade V, Cosgrove R, Fairbairn A, Florin SA et al (2021)
Reimagining the relationship between Gondwanan forests and Aboriginal land management in
Australia’s “Wet Tropics”. iScience. 24(3):102190. https://doi.org/10.1016/j.isci.2021.102190
8. Lourandos H, David B (1998) Comparing long-term archaeological and environmental trends:
North Queensland, arid and semi-arid Australia. The Artefact. 21:105–114
9. Hill R, Griggs P, Incorporated BBN (2000) Rainforest, agriculture and Aboriginal fire-regimes.
Aust Geogr Stud. 38:138–157
10. Pascoe B (2018) Dark Emu: Aboriginal Australia and the birth of agriculture. Magabala Books
11. WTMA. Wet Tropics Management Authority (WTMA) (n.d.) Wet tropics cultural landscape:
rainforest Aboriginal people. Retrieved on March 2, 2022 from https://www.wettropics.gov.
au/our-cultural-landscape
12. Wet Tropics Management Authority (WTMA) (n.d.) Wet tropics cultural landscape: rain-
forest Aboriginal people. Retrieved on March 2, 2022 from https://www.wettropics.gov.au/
our-cultural-landscape
13. Anonymous. Tropical Indigenous Ethnobotany Centre. Accessed on February 12, 2022 from
https://www.tiec.org.au/about-tiec
14. Cock IE (2011) Medicinal and aromatic plants—Australia. In: Ethnopharmacology Section,
Biological, Physiological and Health Sciences, Encyclopedia of Life Support Systems
(EOLSS), Developed under the Auspices of the UNESCO. EOLSS Publishers, Oxford.
Available online: http://www.eolss.net. Accessed 30 Apr 2022
15. Wet Tropics Management Authority. Australia’s tropical rainforests world heritage: Fact sheet-
bush medicine. Accessed on February 13, 2022 from https://www.wettropics.gov.au/site/user-
assets/docs/bushmedicine.pdf
16. Webb LJ (1969) The use of plant medicines and poisons by Australian aborigines.
Mankind. 7:137
17. Roth W (1903) Superstition, magic, and medicine, North Queensland Ethnography Bulletin
No. 5. Government Printer, Brisbane
18. Tithi NA (2017) Chemical and biological investigations of the Yaegl medicinal plant Alphitonia
excelsa. Accessed on February 28, 2022 http://hdl.handle.net/1959.14/1267484
19. Fuentes RG, Valenciano AL, Cassera MB, Kingston DGI (2020) Antiproliferative and
antiplasmodial investigation of Alphitonia excelsa and Arcangelesia flava. Philipp J Sci
149(1):115–120
20. Lassak EV, McCarthy T (2011) Australian medicinal plants: a complete guide to identification
and usage. Reed New Holland, Chatswood
21. FloraNT: Nothern territory flora online. Accessed on February 14, 2022 from http://eflora.
nt.gov.au/home
376 K. Yeshi and P. Wangchuk
45. Griffin WJ, Phippard JH (1971) Alkaloids of the leaves of Erythrophleum chlorostachys.
Phytochemistry. 10:2793–2797
46. Falkiner MJ, Faux AF, Loder JW, Nearn RH (1975) Isolation of unstable alkaloids from
Erythrophleum chlorostachys (Leguminosae). Aust J Chem. 28:645–650
47. Yeshi K, Turpin G, Wangchuk P. Anti-inflammatory activities of medicinal plants from
Mbabaram community of the Far North Queensland (unpublished data). 2020.
48. Akter K, Barnes EC, Brophy JJ, Harrington D, Community Elders Y, Vemulpad SR et al (2016)
Phytochemical profile and antibacterial and antioxidant activities of medicinal plants used by
Aboriginal people of New South Wales, Australia. Evid Based Complement Alternat Med.
2016:4683059. https://doi.org/10.1155/2016/4683059
49. Maiden JH (1913) Appendix: Aboriginal method of obtaining water. In: The Forest Flora of
New South Wales, vol 6, Part LI. NSW Government Printer, Sydney
50. Anderson DMW, Bell PC, McNab CGA (1972) Analysis of six Acacia gum exudates of the
series phyllodineae. Phytochemistry 11:1751–1754
51. Webb LJ (1959) Proc Royal Soc Queensland. 71:103
52. Vigil de Mello SV, da Rosa JS, Facchin BM, Luz AB, Vicente G, Faqueti LG et al (2016)
Beneficial effect of Ageratum conyzoides Linn (Asteraceae) upon inflammatory response
induced by carrageenan into the mice pleural cavity. J Ethnopharmacol. 194:337–347. https://
doi.org/10.1016/j.jep.2016.09.003
53. Adesogan EK, Okunade AL (1979) A new flavone from Aegeratum conyzoides. Phytochemistry.
18(11):1863–1864
54. Moreira MD, Picanco MC, Barbosa LC, Guedes RN, Barros EC, Campos MR (2007)
Compounds from Ageratum conyzoides: isolation, structural elucidation and insecticidal
activity. Pest Manag Sci. 63(6):615–621. https://doi.org/10.1002/ps.1376
55. Vyas AV, Mulchandani NB (1984) Structure reinvestigation of Conyzorigun, a new chromone
from Ageratum conyzoides. J Chem Soc Perkin Trans. 1:2945–2947
56. Bosi CF, Rosa DW, Grougnet R, Lemonakis N, Halabalaki M, Skaltsounis AL et al (2013)
Pyrrolizidine alkaloids in medicinal tea of Ageratum conyzoides. Revista Brasileira de
Farmacognosia. 23(3):425–432. https://doi.org/10.1590/s0102-695x2013005000028
57. Nour AM, Khalid SA, Kaiser M, Brun R, Abdalla WE, Schmidt TJ (2010) The antiproto-
zoal activity of methylated flavonoids from Ageratum conyzoides L. J Ethnopharmacol.
129(1):127–130. https://doi.org/10.1016/j.jep.2010.02.015
58. Vyas AV, Mulchandani NB (1986) Polyoxygenated flavones from Aegeratum conyzoides.
Phytochemistry. 25(11):2625–2627
59. Cribb AB, Cribb JW (1981) Wild medicine in Australia. Collins, Sydney
60. Webb LJ (1959) The use of plant medicines and poisons by Australian Aborigines. Mankind.
7:137–172
61. Simpson BS, Claudie DJ, Smith NM, Gerber JP, McKinnon RA, Semple SJ (2011) Flavonoids
from the leaves and stems of Dodonaea polyandra: a Northern Kaanju medicinal plant.
Phytochemistry. 72(14-15):1883–1888. https://doi.org/10.1016/j.phytochem.2011.05.006
62. Simpson B, Claudie D, Smith N, Wang J, McKinnon R, Semple S (2010) Evaluation of
the anti-inflammatory properties of Dodonaea polyandra, a Kaanju traditional medicine. J
Ethnopharmacol. 132(1):340–343. https://doi.org/10.1016/j.jep.2010.07.012
63. Maiden JH (1889) The useful native plants of Australia. Turner & Henderson, Sydney
64. Gnanaraj C, Shah MD, Makki JS, Iqbal M (2016) Hepatoprotective effects of Flagellaria indica
are mediated through the suppression of pro-inflammatory cytokines and oxidative stress mark-
ers in rats. Pharm Biol. 54(8):1420–1433. https://doi.org/10.3109/13880209.2015.1104697
65. Williams C (2011) Medicinal plants in Australia. Volume II gums, resins, tannin and essential
oils. Rosenberg Publishing Pty Ltd., Kenthurst
66. Rasmussen M, Ridley DD, Ritchie E, Taylor WC (1968) Chemical studies of proteaceae
III. The structure determination and synthesis of striatol, a novel phenol from Grevillea striata
R.Br. Aust J Chem. 21:2989–2999
378 K. Yeshi and P. Wangchuk
67. Roufogalis BD, Li Q, Tran VH, Kable EPW, Duke CC (1999) Investigation of plant -derived
phenolic compounds as plasma membrane Ca2+-ATPase inhibitors with potential cardiovascu-
lar activity. Drug Develop Res 46:235–249
68. Hui WH, Ng KK, Fukamiya N, Koreeda M (1971) Isolation and structure of macarangonol,
a diterpene ketol from Macaranga tanarius. Food Agric Org United Nations 10:1617–1620
69. Kawakami S, Harinantenaina L, Matsunami K, Otsuka H, Shinzato T, Takeda Y (2008)
Macaflavanones A-G, prenylated flavonones from the leaves of Macaranga tanarius. J Nat
Prod. 71:1872–1876
70. Tseng M-H, Chou C-H, Chen Y-M, Kuo Y-H (2001) Allelopathic prenylflavanones from the
fallen leaves of Macaranga tanarius. J Nat Prod. 64:827–828
71. Fletcher TG (2007) Thanakupi’s guide to language & culture: a Thaynakwith dictionary.
Jennifer Isaacs Arts & Publishing, North Sydney
72. Al-Sayed E, Michel HE, Khattab MA, El-Shazly M, Singab AN (2020) Protective role of
casuarinin from Melaleuca leucadendra against ethanol-induced gastric ulcer in rats. Planta
Med. 86(1):32–44. https://doi.org/10.1055/a-1031-7328
73. Brophy JJ (1988) Melaleuca leucodendra L. leaf oil: two phenylpropanoid chemotypes. Flav
Fragr J 3:43–46
74. Hashim AN, Swilam NF, Moustafa ES, Bakry SM, Labib RM, Barakat HH et al (2018) A cyto-
toxic flavonol glycoside from Melaleuca leucadendra leaves extract with immunostimulant
activity. Pharmazie. 73(1):61–64. https://doi.org/10.1691/ph.2018.7785
75. Pino J, Bello A, Urquiola A, Aguero J, Marbot R (2002) Chemical composition of Cajuput
oil (Melaleuca leucadendra L.) from Cuba. J Essential Oil Res 14(1):10–11. https://doi.org/1
0.1080/10412905.2002.9699744
76. Pino JA, Regalado ELR, J.L., Fernández MD. (2010) Phytochemical analysis and in vitro free-
radical-scavenging activities of the essential oils from leaf and fruit of Melaleuca leucadendra
L. Chemistry and Biodiversity. 7(9):2281–2288
77. Lassak EV, McCarthy T (1983) Australian medicinal plants. Methuen Australia
78. Macleod JK, Rasmussen HB (1999) A hydroxy-β-caryophyllene from Pterocaulon serrula-
tum. Phytochemistry. 50(1):105–108. https://doi.org/10.1016/S0031-9422(98)00460-9
79. Siswadi S, Saragih GS (2021) Phytochemical analysis of bioactive compounds in ethanolic
extract of Sterculia quadrifida R.Br. International Conference on Life Sciences and Technology
(ICoLiST 2020)
80. Taebe B, Imrawati I, Rachman CN (2019) Isolation and characterisation of the steroid com-
pounds from aceton extract of Faloak leaves (Sterculia quadrifida R.Br). J Pharm Med Sci 3(2)
81. Liu M, Han J, Feng Y, Guymer G, Forster PI, Quinn RJ (2021) Antimicrobial
benzyltetrahydroisoquinoline-derived alkaloids from the leaves of Doryphora aromatica. J
Nat Prod. 84(3):676–682. https://doi.org/10.1021/acs.jnatprod.0c01093
82. Bick IC, Leo W, Richards M (1980) Alkaloids of Doryphora aromatica. Aust J Chem
33(1):225–228
83. Canter PH, Thomas H, Ernst E (2005) Bringing medicinal plants into cultivation: opportunities
and challenges for biotechnology. Trends Biotechnol. 23(4):180–185
84. Yeshi K, Crayn D, Ritmejeryte E, Wangchuk P (2022) Plant secondary metabolites produced
in response to abiotic stresses has potential application in pharmaceutical product develop-
ment. Molecules. 27(1):1–31. https://doi.org/10.3390/molecules27010313
85. Adegboye O, Field MA, Kupz A, Pai S, Sharma D, Smout MJ et al (2021) Natural-product-
based solutions for tropical infectious diseases. Clin Microbiol Rev 34(4):e0034820. https://
doi.org/10.1128/CMR.00348-20
86. United Nations: Convention on Biological Diversity (1992). http://www.cbd.int/doc/legal/cbd-
en.pdf. Accessed 2021
87. Yeshi K, Kashyap S, Yangdon P, Wangchuk P (2017) Taxonomical identification of himala-
yan edible medicinal plants in Bhutan and the phenolic contents and antioxidant activity of
selected plants. JBAPN. 7(2):89–106. https://doi.org/10.1080/22311866.2017.1325008
Bush Medicinal Plants of the Australian Wet Tropics and Their Biodiscovery Potential 379
88. Nugraha AS, Dayli IR, Sukrisno Putri CPZ, Firli LN, Widhi Pratama AN, Triatmoko B
et al (2022) Isolation of antibacterial depside constituents from Indonesian Folious Lichen,
Candelaria fibrosa. J Biol Active Prod Nature. 12(1):24–32
89. Anonymous. Biodiscovery and bussiness, Queensland Government. Retrieved on February
27, 2022 from https://www.business.qld.gov.au/industries/science-it-creative/science/bio-
discovery/qld#:~:text=The%20Biodiscovery%20Act%202004%20aims,equitable%20
benefit%20to%20the%20community.&text=This%20obligation%20applies%20to%20
biodiscovery,collection%20from%20anywhere%20in%20Queensland
Trillium govanianum – A Promising
Endemic Medicinal Herb of the Himalaya
1 Introduction
Plants have been used for medicinal purposes long before recorded history. Even
today, the traditional medicinal uses of plants play an important role in providing
primary health care [1, 2]. The World Health Organization (WHO) has documented
over 21,000 plant species all over the world used as medicinal [3]. About 70–80% of
people around the globe rely on medicinal plants for primary healthcare and liveli-
hood [4]. About 3.5–4 billion people in the world are dependent on plants as potent
sources of drugs [5]. However, simultaneously, the recent increase in market demand
for herbal products has resulted in huge pressure on natural populations of medicinal
plants, which in turn has brought many prized species to the brink of extinction [6–8].
A systematic review of medicinal plant species especially with a detailed focus
on their botanical, ethnomedicinal, phytochemical, and pharmacological aspects
effectively integrates the precious information but scattered in research studies [9].
The reviews critically synthesize the disparate research knowledge at a single scien-
tific platform and help researchers to assess the progress achieved so far, highlight
knowledge gaps and suggest future research directions. In recent times, reviews on
several medicinal plant species with a focus on their ethnomedical use, phytochem-
istry and pharmacology have been published [10]. These critical review help in
linking the novel leads from ethnomedicinal uses of plant species to active phyto-
chemical constituents and pharmacological properties. These studies show that the
use of medicinal plant products has been growing not only as therapeutically active
agents, but also unlock the rich reservoir of naturally occurring lead compounds in
drug discovery [11].
Trillium govanianum Wall. ex D.Don (English Name: Himalayan Trillium) has
recently emerged as one of the most important medicinal species of the Himalayan
region with a high therapeutic value [12]. The market demand for the raw material
of this medicinal plant has increased manifold owing to its high concentration of
diosgenin; and the quantity of the diosgenin is almost threefold higher than reported
from other plant sources: Asparagus, Chlorophytum, Dioscorea and Trigonella spe-
cies [13]. Diosgenin has many pharmaceutical uses: it is used as anti-cancerous,
anti-ageing, anti-inflammatory and precursor for manufacturing many steroidal
drugs [14, 15]. However, the collection of this medicinal plant species to meet the
rising demand of the pharmaceutical industry, and more worryingly its unsustain-
able harvesting from the wild, is alarmingly depleting its natural populations across
the Himalaya. The species is almost threatened now in the entire Himalayan region
[7]. Its slow life cycle taking several years to reach the flowering stage, specific
habitat requirement, low population density, no commercialization alternatives, her-
bivory and overharvesting for pharmaceutical industries has brought this species to
the threatened status [16].
It is in this background that the present review provides a comprehensive synthe-
sis of the research studies conducted on the botanical, ethnomedicinal, phytochemi-
cal and pharmacological aspects of T. govanianum. Hopefully, this scientific
synthesis on T. govanianum will help in stocktaking the research progress achieved
so far and prospects of future research opportunities on this promising endemic
medicinal plant from the Himalaya.
The published literature on T. govanianum were retrieved using popular and rele-
vant science search engines and database including scientific journals, books, theses
and electronic search (Google Scholar, Science Direct, Research Gate, JSTOR and
PubMed). We used the following keywords: “Trillium”, “Trillium govanianum”,
“T. govanianum”, “Himalayan Trillium” independently, and combined with other
terms: “botany”, “morphology”, “taxonomy”, “phenology”, “biogeography”, “tra-
ditional uses”, “ethnic uses”, “ethnomedicine”, “local uses”, “ethnomedicinal uses”,
“phytochemistry”, “steroids”, “saponins”, “steroidal saponins”, “active com-
pounds”, “chemical constituents”, “pharmacology”, “biological activity”, “activ-
ity”, “toxic”, “toxicity”, “propagation”, “threat” and “conservation”. In total, 54
studies spanning from the year 1994 to April 2020 were critically read, evaluated
and incorporated in this review (Fig. 1).
The chemical structures and formulae of compounds have been sourced and vali-
dated by using the PubChem database. Chemical structures were drawn using Chem
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 383
2.1 Botany
2 whorls, shorter than the perianth; filaments ca. 4 mm long; anthers basifixed,
4–5 mm long, curved, and dehiscence longitudinal. Ovary superior, purple-red,
ovoid-globose, 3-loculed and ovules few to many per locule. Fruit red, globose
berry, 1–2 cm in diameter, seeds numerous, oblong, ca. 2.5 mm long, with a pulpy
lateral appendage (Fig. 2).
Fig. 2 Trillium govanianum. (a) Habitat, (b) Habit, (c) Rhizome, Individual plants with (d) one
leaf, (e) two leaves, (f) threes leaves, (g) Bisexual flower, (h) Dark purple tepals (i) Berry fruit
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 385
2.2 Biogeography
The genus Trillium, with ca. 49 species world over, is one of the representative ele-
ments of the northern temperate forests. It has been reported that the genus origi-
nated in Arcto-Tertiary and shows geographical distribution between North America
and eastern Asia including Japan [52, 53]. About 40 species occur in North America,
the remaining nine species are found in Asia [54]. T. govanianum (Himalayan
Trillium), one of the Asian species, is endemic to the Himalayan region. It is distrib-
uted in temperate and subalpine zones of the Himalaya at altitudes ranging from
1800 to 3500 m (m.s.l.), especially in Afghanistan, Pakistan, India, Bhutan, Nepal
and China (Tibet) [55, 56]. In the Indian Himalaya, this species is primarily distrib-
uted in the western region (Jammu and Kashmir, Himachal Pradesh, Uttarakhand)
and is less common in the eastern region [16]. The species usually prefers moist
shady areas in forest understory for its profuse growth [57, 58].
T. govanianum is a typical early spring geophyte. It grows each year in late March
after snow melts, flowering starts usually in April and fruiting season lasts from
May to June. Leaf senescence occurs in July/August at lower altitudes and
September/October at higher altitudes. During the flowering phase, both vegetative
and flowering individuals can be found on the forest floor. The vegetative individu-
als comprise three morphologically distinct forms: individuals bearing 1-leaf,
2-leaves and 3-leaves (Fig. 2). As the plant shows a relatively slower growth rate,
taking many years to enter the reproductive phase, the life span of the species is
reported to be ca. 30 years [16, 59].
Plant tissue culture refers to the culturing of different plant explants such as leaves,
seeds, tissues, cells or protoplasts on a chemically defined synthetic nutrient media
under sterile and controlled conditions of light, temperature, humidity, etc. [60].
The in vitro multiplication technique has an advantage over in vivo conditions, as it
can yield a definite and uniform production system of quality and yield [61, 62]. In
fact, in contrast to the time-consuming indigenous techniques, micro-propagation
has led to the easy generation of somatic clones (ramets) of selected genotypes [63].
T. govanianum, being a rich treasure of novel phytochemicals, has led to its overex-
ploitation from its natural habitats rendering it endangered [12]. Therefore, plant
tissue culture offers an important and potential alternative method for rapid multi-
plication and germplasm conservation of this medicinally important plant species.
386 K. Rashid et al.
2.5 Ethnomedicinal Uses
T. govanianum has been widely used in traditional medicinal systems because of its
broad therapeutic properties. It is commonly known as “tri pater”, “nag chhatri” or
“chotasatwa” in India, “teenpatra” or “matarzela” in Pakistan, and “Xizangyan ling
cao” in Tibet (China). The underground part of the plant, i.e. rhizome is a prime raw
material used in trade. The plant is used in several traditional medicine systems and
used for the treatment of ailments such as boils, dysentery, inflammation, menstrual
disorders, sex-related disorders, wounds, as an antiseptic, and for improving general
health [18, 20, 25, 65–67]. It has been reported that the powdered rhizome is used
as body and sexual tonic [28]. In Jammu and Kashmir (India), the Gujjar and
Bakerwal tribes make tea from dried rhizomes to cure inflammation and kidney
diseases [17]; and fresh rhizome is consumed with herbal tea to treat appetite disor-
der [33]. In Himachal Pradesh (India), tribal communities take dried form of rhi-
zome with cow milk in early morning to cure menstrual and reproductive disorder
[18]. In Shimla district of Himachal Pradesh people use leaves and root paste to cure
minor cuts, wounds, and foot cracks [35]. The migratory shepherds in Kinnaur dis-
trict in Himachal Pradesh use dried leaves and root powder for curing cough, fever,
vomiting [37], and the juice of fresh leaves and roots is used for the treatment of
body pain, internal injury and foot and mouth diseases [38]. In Azad Jammu and
Kashmir (Pakistan), dried root powder is applied externally to cure wounds [19].
Besides human diseases, the leaves of this plant are used for treating indigestion and
parasites in livestock [28]. In the Bandipora area of Jammu and Kashmir (India), the
crushed rhizome is given in the form of balls to livestock against worms [21]. The
summary of the reported ethnopharmacological uses of different parts of T. gova-
nianum is presented in Table 1.
2.6 Phytochemistry
Table 1 (continued)
Plant part used/ Local name(s)/
S. No. Diseases Mode of use Place Reference(s)
9. Body and sexual tonic Rhizome/ Tandhijarri/ [28]
Powdered form used Saifalmaluk Lake,
as body and sexual Naran Valley, Pakistan
tonic.
10. Arthritis Rhizome/ Nag chhatri/ [29]
Dried powder along Churah subdivision,
with buttermilk is District Chamba,
used Himachal Pradesh,
India
11. Rheumatism, fever, and Rhizome/ ChhotaSatuwa, Nag [30]
sexual disorder chhatri/
Powder or paste is Uttarkashi District,
given with warm Uttarakhand, India
water
12. Body weakness Rhizome/ Nag chhatri/ [31]
Rhizome are boiled North western
in milk and taken Himalaya, Jammu and
orally Kashmir, India
13. Impotency; tumor Root/ Tripatra/ [32]
Dried powder is used Bheri, Muzaffarabad,
Pakistan
14. Appetite disorder Rhizome/ Sheethkhar/ [33]
Fresh rhizomes Bandipora District,
consumed in herbal Jammu and Kashmir,
tea India
15. Tonic and emetic Rhizome/ Matarjarai/ [34]
Oral administration Hindukush Range,
of watery extract of District Swat, Pakistan
rhizome.
16. Wounds and cut healing Rhizome, Leaves/ Nag chhatri/Satwa [35, 36]
Fresh leaves or dry Chopal region, District
rhizome paste and Shimla, Himachal
slurry is applied Pradesh, India
externally on
wounds, minor cuts
and foot cracks till it
heals
17. Cough; fever; vomiting Leaves, Rhizome/ Nag chhatri/ [37]
Oral application of Rakchham area,
dried powder of District Kinnaur,
leaves and rhizome Himachal Predesh,
India
18. Body pain; internal Leaves, Rhizome/ Nag chhatri, [38]
injury The juice of fresh Baspa valley, District
leaves and rhizome Kinnaur, Himachal
are given orally Predesh, India
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 389
Fig. 3 Structure of phytochemicals isolated and identified from Trillium govanianum: Unsaturated
fatty acids (1–12); Saturated fatty acids (13–19); Phenolics (20–23); Steroids (24–29)
392 K. Rashid et al.
2.7 Pharmacology
The crude extracts as well as pure compounds obtained from T. govanianum have
been used to validate the ethnomedicinal uses and further assessed for novel bio-
logical activities. The key bioactive constituents from the species have been recently
subjected to various pharmacological investigations and this has revealed promising
antibacterial, antifungal, anticancer, antifertility contraceptive, analgesic, anti-
inflammatory, antioxidant and anti-parasitic properties of the species.
2.7.1 Antibacterial Activity
was found to be more effective against S. aureus at all the concentrations as com-
pared to E. coli and Y. pestis in three solvents: ethanol, methanol and acetone; and
in case of distilled water rhizome extract, Y. pestis was found to be more effective as
compared to S. aureus and E. coli. In another study [40], antibacterial activity of the
MeOH extract of rhizome of T. govanianum and its solid phase extractions (SPE)
was tested on three gram-positive bacteria (Staphylococcus aureus, Bacillus subti-
lis, Micrococcus luteus), and two gram-negative bacteria(Escherichia coli,
Klebsiella oxytoca). Using Resazurin microtiter plate assay method, higher
398 K. Rashid et al.
antibacterial activity was detected against gram-positive bacteria. In case of its SPE
fraction TGMF1, the most significant activity was detected against M. luteus (MIC:
0.156 mg/mL). The SPE fraction TGMF2 showed the most significant antibacterial
activity against B. subtilis and M. luteus (MIC: 0.615 and 1.25 mg/mL). The SPE
fraction TGMF3 showed the most significant antibacterial activity against B. subti-
lis, M. luteus and S. aureus (MIC: 0.0195, 0.039 and 0.31 mg/mL). The SPE frac-
tion TGMF4 exhibited a notable antibacterial activity against S. aureus, B. subtilis
and M. luteus (MIC: 0.615, 0.039 and 0.156 mg/mL) and showed no activity against
E. coli and K. oxytoca (MIC: 1.25 and 0.615 mg/mL).
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 399
2.7.2 Antifungal Activity
2.7.3 Anticancer Activity
Khan et al. [67] evaluated the cytotoxic potential of methanolic extracts of the rhi-
zomes of T. govanianum and its solid phase extraction (SPE) fractions and also their
application on the breasts (MCF7), liver (HepG2), lungs (A549) and urinary bladder
(EJ138) cancer cell lines using in vitro MTT (3-(4, 5-dimethylthiazol-2-yl)-2,
5-diphenyltetrazolium bromide) cytotoxicity/viability assay, demonstrated their
concentration-dependent activity. The MeOH extract displayed the highest level of
cytotoxicity against the urinary bladder cell line (EJ138; IC50 = 5 μg/mL). The SPE
fraction TGMF1 showed the most significant cytotoxicity against EJ138 (IC50 = 6 μ g/
mL). The SPE fraction TGMF2 showed the most prominent cytotoxicity against the
lung cancer cell line A549 (IC50 = 6 μg/mL) and the SPE fraction TGMF3 showed
the most significant cytotoxicity against the EJ138 cell line (IC50 = 9 μg/mL). The
SPE fraction TGMF4, which contained the least polar components of the parent
MeOH extract, exhibited notable cytotoxicity against the EJ138 cell line
(IC50 = 13 μg/mL). In another study, Sharma et al. [75] showed cytotoxicity of
400 K. Rashid et al.
rhizome and hydroalcoholic extract which were carried out by the MTT (3-(4,
5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay on three cell lines:
MDCK, MCF-7 and MDA-MB-23. The results indicated that hydrolyzed extract
showed less toxicity in MDCK (normal cell line) cells but significantly reduced the
proliferation of MCF-7 and MDA-MB-231 cancer cells. Rehman et al. [39] studied
the cytotoxic activity of methanol extract and its fractions against two cancer cell
lines, HeLa (cervical cancer cells) and PC-3 (prostate cancer cells) by using the
MTT(3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide) assay. In
this study, based on IC50 (μg/mL) values, chloroform (0.8 ± 0.2), ethyl acetate
(1.4 ± 0.1) and butanol (1.6 ± 0.3) fractions exhibited anti-proliferative activity on
HeLa cells in comparison to anticancer drug doxorubicin (0.3 ± 0.0). Similarly, the
chloroform fraction was effective against PC-3 cells while ethyl acetate and butanol
fractions were least effective.
Being a rich source of steroids, T. govanianum has been shown to possess antifertil-
ity properties. The anti-implantation activity of T. govanianum rhizome extract was
performed in female wistar rat with proven fertility and its estrogenic/anti-estrogenic
effect was evaluated in ovariectomized females. A dose-dependent anti-implantation
effect and a significant increase in uterus weight were observed. It was observed
that, at 250 mg/kg dose, 100% inhibition of implantation was found. This anti-
implantation effect was attributed to endometrial thickening and changes in the
estrogenic profile [76].
The inflammation produces reactive oxygen species (ROS), which in turn further
perpetuate inflammation. The ROS produced during the inflammatory process
causes tissue damage [77]. The production of highly reactive species such as ROS,
reactive nitrogen species (RNS) and hypochlorous acid (HOCl) at the site of inflam-
mation exacerbates tissue damage as well as the inflammatory cascade [78, 79].
This ROS-induced tissue damage forms the basis of the pathogenesis of many
chronic inflammatory diseases [80].
The anti-inflammatory and analgesic properties of extracts of T. govanianum
have been evaluated in animal models to confirm its pharmacological properties.
Both in vivo and in vitro anti-inflammatory activity have been determined through
carrageenan-induced paw edema assay and luminol-enhanced chemiluminescence
assay, respectively. The in vivo results demonstrated significant anti-inflammatory
activity possibly by inhibiting release of iNOS, COX2 and inhibiting the production
of tissue necrosis factor (TNF), cytokines (IL-1, IL-2) and nitric oxide synthase
(NOS) release associated with inflammation. This anti-inflammatory activity of
T. govanianum may provide a new path in pharmacological development. The in
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 401
vitro study also depicted anti-inflammatory activities associated with the quenching
of reactive oxygen species or inhibiting the enzymes involved in their production.
The isolated compounds such as pennogenin showed a significant in vitro inhibitory
activity by controlling the release of ROS with an IC50 of 5.0 ± 0.8 μg/mL while
borassoside E and diosgenin showed a weaker inhibition activity. The in vivo anal-
gesic activity was assayed in chemical and thermal-induced nociceptive pain mod-
els. The methanol extract and its solvent fractions showed analgesic responses,
which were exhibited by significant tonic visceral chemical and acute phasic ther-
mal nociception. Therefore, the rhizomatous part of this plant species could act as a
potential and novel source of compounds, which are analgesic and anti-
inflammatory [81].
Free radicals being highly reactive species have the potential to cause oxidative
damage to life-essential molecules such as lipids, proteins and nucleic acids, which
eventually leads to atherosclerosis, ageing, cancer and several other diseases in
humans [82–84]. Besides the antioxidant vitamins, a number of several other com-
pounds such as vitamins E, C, beta-carotene and folic acid have been reported to
have strong antioxidant potential [85]. There are continuous research efforts being
made to search for new and elite compounds and the identification of food ingredi-
ents with an antioxidant potential [86].
The in vitro antioxidant activity of methanol extracts and its fractions (EtOAc.Fr,
CHL.Fr, BuOH.Fr, Hex.Fr) of T. govanianum rhizome was determined by the DPPH
(1, 1-diphenylpicryl-1-picrylhydrazyl) free radical scavenging assay. This activity
was analyzed against the synthetic antioxidants BHT and ascorbic acid. The results
have shown that a higher scavenging capacity was measured in Hex-Fr and CHL-Fr
when compared with other fractions. This was due to the presence of certain anti-
oxidant fatty acids (9, 12-octadecadienoic acid and hexadecanoic acid) in n-Hex-fr,
and glycosides, saponins and flavonoids in CHL-fr. However, the antioxidant poten-
tial of the MeOH extract as well as its successive solvent soluble fractions was
lower than some synthetic antioxidants BHT (butylated hydroxytoluene) and ascor-
bic acid. This low scavenging property might be due to the presence of large-size
fatty acids [39]. In another study [70], the antioxidant potential of methanolic
extracts of leaf, stem and rhizome of T. govanianum was determined using
1,1-diphenyl-2-pycrylhydrazyl (DPPH); 2, 20–azinobis, 3-ethylbenzothiazoline-6-
sulfonic acid (ABTS) and ferric-reducing antioxidant power (FRAP) free radical
scavenging assay. It was shown that DPPH, FRAP, activities in the methanolic
extract of rhizome were maximum, whereas ABTS activity was maximum in the
methanolic extract of leaf. IC50 values were found maximum for rhizome (0.15 mg/
ml) followed by leaf (0.165 mg/ml) and least for stem (0.192 mg/ml). Therefore, the
rhizome part was having stronger antioxidant potential than leaf and stem.
402 K. Rashid et al.
2.7.8 Anti-leishmanial Activity
2.7.9 Cytotoxic Activity
Cytotoxicity potential of the MeOH extract of rhizome of T.govanianum and its SPE
fractions was determined through brine shrimp lethality assay, and concentration-
dependent lethality was observed. In this cytotoxicity assay, the LC50 value of meth-
anolic extract was 38.5 μg/mL which was most toxic, while LC50 values of its SPE
fractions TGMF1, TGMF2, TGMF3 and TGMF4 were 40.5, 189.2, 105.6 and
66.5 μg/mL, respectively. The doxorubicin, as a positive control, showed an LC50
value of 1.98 μg/mL [42].
The protein kinase inhibition assay of the MeOH extract of rhizome of T.govanianum
and its SPE was performed by observing hyphae formation in purified isolates of
Streptomyces 85E strain. The effect of MeOH extract and its SPE fractions was
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 403
assessed through the formation of different levels of protein kinase inhibition zones.
The results revealed distinguishable protein kinase inhibitory activity against
Streptomyces 85E strain with 18 mm bald phenotype. This effect might be attributed
to the presence of quercetin which has been reported to inhibit multiple kinases,
thus playing an important role in cancer cell biology [42].
3 Conclusions
should be prioritized for effective and sustainable utilization of the plant at com-
mercial scale. Therefore, for the proper conservation and management of this pre-
cious plant species, it is incumbent to carry out conservation-focused research as
well. Since there is no scientific breakthrough in its commercial farming and pro-
duction, the review recommends further research toward its ex situ propagation and
in vitro regeneration, so as to reduce pressure on the natural populations. It is also
recommended that public awareness and sensitization campaigns should be carried
out to prevent the destructive harvesting by local people in natural habitats across
the Himalaya.
References
1. Ganie AH, Tali BA, Shapoo GA, Nawchoo IA, Khuroo AA (2019) Ethno-survey of traditional
use of plants as aphrodisiacs in Kashmir Himalaya, India. J Herb Med 17:100256
2. Ganie AH, Tali BA, Nawchoo IA, Khuroo AA, Reshi ZA, Dar GH (2020) In: Dar GH, Khuroo
AA (eds) Biodiversity of the Himalaya: Jammu and Kashmir State. Springer, Singapore,
pp 545–563
3. WHO World Health Organization (2002) Traditional medicine strategy, p. 11
4. Singh JS (2002) The biodiversity crisis: a multifaceted review. Curr Sci 82:638–647
5. Patwardhan B, Warude D, Pushpangadan P, Bhatt N (2005) Ayurveda and traditional Chinese
medicine: a comparative overview. Evid Based Complement Alternat Med 2:465–473
6. Uniyal SK, Singh KN, Jamwal P, Lal B (2006) Traditional use of medicinal plants among the
tribal communities of Chhota Bhangal, Western Himalaya. J Ethnobiol Ethnomed 2:14. https://
doi.org/10.1186/1746-4269-2-14
7. Tali BA, Khuroo AA, Ganie AH, Nawchoo IA (2019a) Diversity, distribution and traditional
uses of medicinal plants in Jammu and Kashmir (J&K) state of Indian Himalayas. J Herb Med
17:100280
8. Hamid M, Khuroo AA, Ahmad R, Rasheed S, Malik AH, Dar GH (2020) In: Dar GH, Khuroo
AA (eds) Biodiversity of the Himalaya: Jammu and Kashmir State. Springer, Singapore,
pp 957–995
9. Dhiman B, Sharma P, Pal PK (2020) Biology, chemical diversity, agronomy, conservation
and industrial importance of Valeriana jatamansi: a natural sedative. J Appl Res Med Aroma
16:100243
10. Rathore S, Debnath P, Kumar R (2020) Kuth {Saussurea costus (Falc.) Lipsch.}: a critically
endangered medicinal plant from Himalaya. J App Res Med Aro Plants:100277
11. Tali BA, Khuroo AA, Nawchoo IA, Ganie AH (2019b) Prioritizing conservation of medici-
nal flora in the Himalayan biodiversity hotspot: an integrated ecological and socioeconomic
approach. Environ Conserv 46:147–154
12. Vidyarthi S, Samant SS, Sharma P (2013) Dwindling status of Trillium govanianum Wall.
ex. D. Don- A case study from Kullu district of Himachal Pradesh, India. J Med Plants Res
7:392–397
13. Singh G, Singh P, Bhandawat A, Singh G, Parmar R, Seth R, Sharma RK (2017) Spatial tran-
scriptome analysis provides insights of key gene(s) involved in steroidal saponin biosynthesis
in medicinally important herb Trillium govanianum. Sci Rep 7:45295. https://doi.org/10.1038/
srep45295
14. Raju J, Rao CA (2012) Diosgenin, a steroid saponin constituent of yams and fenugreek:
emerging evidence for applications in medicine. Bio Com Phytomed:125–142
15. Chaudary S, Chikara SK, Sharma MC, Chaudary AA, Alam Syed B, Chaudary PS, Mehta
A, Patel M, Gosh A, Iriti M (2015) Elicitation of diosgenin production in Trigonella foenum-
graecum (Fenugreek) seedlings of methyl jasmonate. Int J Mol Sci 16:29889–29899
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 405
16. Chauhan HK, Bisht AK, Bhatt ID, Bhatt A, Gallacher D, Santo A (2018) Population change
of Trillium govanianum (Melanthiaceae) amid altered indigenous harvesting practices in the
Indian Himalayas. J Ethnopharmacol 213:302–310
17. Shah A, Bharati KA, Ahmad J, Sharma M (2015) New ethnomedicinal claims from Gujjar
and Bakerwals tribes of Rajouri and Poonch districts of Jammu and Kashmir, India. J
Ethnopharmacol 166:119–128
18. Rani S, Rana J, Rana P (2013) Ethnomedicinal plants of Chamba district, Himachal Pradesh,
India. J Med Plant Res 7:3147–3157
19. Mahmood A, Mahmood A, Malik RN (2012) Indigenous knowledge of medicinal plants from
Leepa valley, Azad Jammu and Kashmir. Pakistan J Ethnopharmacol 143:338–346
20. Sharma P, Samant S (2014) Diversity, distribution and indigenous uses of medicinal plants
in Parbati valley of Kullu district in Himachal Pradesh, Northwestern Himalaya. Asian J Adv
Basic Sci 2:77–98
21. Lone PA, Bhardwaj AK, Shah KW, Tabasum S (2014) Ethnobotanical survey of some threat-
ened medicinal plants of Kashmir Himalaya, India. J Med Plant Res 8:1362–1373
22. Lone PA, Bhardwaj AK, Bahar FA (2013) Traditional knowledge on healing properties of
plants in bandipora district of Jammu and Kashmir, India. Int J Recent Sci Res 4:1755–1765
23. Bhardwaj AK, Lone PA, Dar M, Parray JA, Shah KW (2013) Ethnoveterinary medicinal uses
of plants of district Bandipora of Jammu and Kashmir, India. Int J Trad Nat Med 2:164–178
24. Wagay NA (2016) Ethnobotany from North Kashmir: a review. Life Sci Leafl 80:38–60
25. Ur Rahman S, Ismail M, Khurram M, Haq IU (2015) Pharmacognostic and ethnomedicinal
studies on Trillium govanianum. Pakistan J Bot 47:187–192
26. Lone PA, Bhardwaj AK, Bahar FA (2015) Study of indigenous/traditional medicinal plant
knowledge- an Endeavour towards new drug discovery. Afr J Tradit Complement Altern Med
12:73–95
27. Tabani AA, Khan SM, Majid A, Hussain I, Saeed M, Khan SA, Naveed K, Ali S (2016)
Ethnomedicinal studies of plants from Shangla district with special reference to economically
important species. ARPN J Agri Biol Sci 11:223–229
28. Khan SM, Page S, Ahmad H, Shaheen H, Ullah Z, Ahmad M, Harper DM (2013) Medicinal
flora and ethnoecological knowledge in the Naran Valley, Western Himalaya, Pakistan. J
Ethnobiol Ethnomed 9:4–16
29. Rana D, Bhatt A, Lal B (2019) Ethnobotanical knowledge among the semi-pastoral Gujjar tribe
in the high altitude (Adhwari’s) of Churah subdivision, district Chamba, Western Himalaya. J
Ethnobiol Ethnomed 15:10. https://doi.org/10.1186/s13002-019-0286-3
30. Nand K, Naithani S (2018) Ethnobotanical uses of wild medicinal plants by the local commu-
nity in the Asi Ganga sub-basin, Western Himalaya. J Complement Med Res 9:34–46
31. Dutt HC, Bhagat N, Pandita S (2015) Oral traditional knowledge on medicinal plants injeop-
ardy among Gaddi shepherds in hills of northwestern Himalaya, J&K, India. J Ethnopharmacol
168:337–348
32. Ahmed MJ, Akhtar T (2016) Indigenous knowledge of the use of medicinal plants in Bheri,
Muzaffarabad, Azad Kashmir, Pakistan. Eur J Integr Med 8:560–569
33. Singh B, Sultan P, Hassan QP, Gairola S, Bedi YS (2016) Ethnobotany, traditional knowledge,
and diversity of wild edible plants and fungi: a case study in the Bandipora District of Kashmir
Himalaya, India. Int J Geogr Inf Syst 22:247–278
34. Ali A, Badshah L, Hussain F (2018) Ethnobotanical appraisal and conservation status of
medicinal plants in Hindukush Range, District Swat, Pakistan. Int J Geogr Inf Syst 24:332–355
35. Singh J, Singh J, Kumar N, Jishtu V, Sharma S, Dhupper R (2017a) Ethno-medicinal plants
used by indigenous people of Kanda range, Chopal forest division, Himachal Pradesh. World
J Pharm Pharm Sci 7:697–710
36. Singh J, Singh J, Sharma D (2018) Traditional wisdom to treat the most common ailments in
Chopal region of Shimla district, Himachal Pradesh, India. Plant Arch 18:2759–2769
37. Radha PS (2019) Assessment of wild medicinal plant used by migratory shepherds in alpine
area of Rakchham-Chitkul wild life sanctuary of district Kinnaur in Himachal Pradesh. Plant
Arch 19:418–429
406 K. Rashid et al.
38. Radha PS, Saha S (2020) Documenting traditional wisdom before they are forgotten: a study on
the ethnoveterinary uses of mountain plants among the trans-Himalayan Migratory Shepherds
in the Kinnaur District of Himachal Pradesh, India. Preprints 2020:2020010343
39. Ur Rahman S, Ismail M, Shah MR, Iriti M, Shahid M (2015c) GC/MS analysis, free radical
scavenging, anticancer and glucuronidase inhibitory activities of Trillium govanianum rhi-
zome. Bangladesh J Pharmacol 10:577–583
40. Khan KM, Sarker SD, Khan GA, Saleem H, Khan AS, Mannan A (2019) Phytochemical pro-
filing and evaluation of modified resazurin microtiter plate assay of the roots of Trillium gova-
nianum. Nat Prod Res:1–5. https://doi.org/10.1080/14786
41. Ur Rahman S, Adhikari A, Ismail M, Shah MR, Khurram M, Anis I, Ali F (2017) A new trihy-
droxylated fatty acid and phytoecdysteroids from rhizomes of Trillium govanianum. Rec Nat
Prod 11:323–327
42. Khan KM, Nahar L, Mannan A, Haq I, Arfan M, Khan GA, Hussain I, Sarker SD (2017)
Cytotoxicity, in vitro anti-Leishmanial and fingerprint HPLC- photodiode array analysis of the
roots of Trillium govanianum. Nat Prod Res 32:2193–2201
43. Ur Rahman S, Ismail M, Shah MR, Adhikari A, Anis I, Ahmad MS, Khurram M (2015b)
Govanoside A, a new steroidal saponin from rhizomes of Trillium govanianum. Steroids
104:270–275
44. WCSP World Checklist of Selected Plant Families, Royal Botanic Gardens, Kew 2013. http://
apps.kew.org/wcsp/qsearch. do
45. Christenhusz MJM, Byng JW (2016) The number of known plants species in the world and its
annual increase. Phytotaxa 261:201–217
46. Zomlefer WB, Williams NH, Whitten WM, Judd WS (2001) Generic circumscriptions and
relationships in the tribe Melanthieae (Liliales, Melanthiaceae), with emphasis on Zigadenus:
evidence from ITS and TRNL-F sequence date. Am J Bot Bot Soc Amer 88:1657–1669
47. Fuse S, Tamura MN (2000) A phylogenetic analysis of the plastid matK gene with emphasis
on Melanthiaceae sensulato. Plant Biol 2:415–427
48. Chase MW, Christenhusz MJM, Fay MF, Byng JW, Judd WS, Soltis DE, Mabberley DJ et al
(2016) An update of the Angiosperm Phylogeny Group classification for the orders and fami-
lies of flowering plants: APG IV. Bot J Linn Soc 181:1–20
49. Fukuda I (2001) The origin and evolution in Trillium.1. The origin of the Himalayan Trillium
govanianum. Cytologia 66:105–111
50. Pellicer J, Kelly LJ, Leitch IJ, Zomlefer WB, Fay MF (2014) A universe of dwarfs and giants:
genome size and chromosome evolution in the monocot family Melanthiaceae. New Phytol
201:1484–1497
51. Chandola V, Nautiyal AR, Chandra S, Kumar RR (2019) A review on prioritizing conservation
of Himalayan medicinal plant species: case of Trillium govanianum (Wall.ex.D.Don) Kunth.
J Med Plants Stud 7:23–27
52. Li HL (1952) Floristic relationships between eastern Asia and eastern North America. Trans
Amer Phil Soc 42:371–424
53. Axelrod DI (1966) Origin of deciduous and evergreen habitats in temperate forests.
Evolution 20:1–15
54. Ohara M, Kawano S (2005) Life-history monographs of Japanese plants. 2: Trillium camschat-
cense Ker-Gawl. (Trilliaceae). Plant Spec Biol 20:75–82
55. Polunin O, Stainton A (1984) Flowers of the Himalaya. Oxford University Press, New Delhi
56. Roskov Y, Abucay L, Orrell T, Nicolson D, Bailly N, Kirk PM, Bourgoin T, DeWalt RE,
Decock W, DeWever A, Nieukerken E, Zarucchi J, Penev L (eds) (2018) Species 2000 &ITIS
Catalogue of Life, 28th March 2018. Digital resource at www.catalogueoflife.org/col. Species
2000: Naturalis, Leiden. ISSN 2405–8858
57. Samant SS, Dhar V, Palni LMS (1998) Medicinal plants of Indian Himalaya: diversity distribu-
tion potential volumes, vol 163. Gyanoday Prakhasan, Nainital
58. Kubota S, Kameyama Y, Ohara M (2006) A reconsideration of relationship among Japanese
Trillium species based on karyology and AFLP data. Plant Syst Evol 261:129–137
Trillium govanianum – A Promising Endemic Medicinal Herb of the Himalaya 407
59. Chauhan HK, Bisht AK, Bhatt ID, Bhatt A, Gallacher D, Santo A (2019) Trillium—toward
sustainable utilization of a biologically distinct genus valued for traditional medicine. Bot Rev
85:252–272
60. Bhojwani SS, Razdan MK (1986) Plant tissue culture: theory and practice. Elsevier, Amsterdam
61. Fay MF (1992) Conservation of rare and endangered plants using in vitro methods. In Vitro
Cell Dev Biol Plant 28:1–4
62. Zhao P, Iwamoto Y, Kouno I, Egami Y, Yamamoto H (2004) Stimulating the production of
homoisoflavonoids in cell suspension cultures of Caesalpinia pulcherrima using cork tissue.
Phytochemistry 65:2455–2461
63. Nin S, Morosi E, Schiff S, Bennici A (1996) Callus cultures of Artemisi absinthium L.: initia-
tion, growth optimization and organogenesis. Plant Cell Tiss Org 45:67–72
64. Chauhan HK, Bisht AK, Bhatt ID, Bhatt A (2020) Protocol for vegetative propagation of
Trillium govanianum Wall ex D. Don. J Appl Res Med 16:100233. https://doi.org/10.1016/j.
jarmap.2019.100233
65. Shah R (2006) Nature’s medicinal plant of Uttaranchal. Gyanodaya Prakhasan, Nainital
66. Pant S, Samant S (2010) Ethnobotanical observations in the Mornaula reserve forest of
Komoun, est Himalaya, India. Ethnobot Leafl 14:193–217
67. Khan KM, Nahar L, Al-Groshi A, Zavoianu AG, Evans A, Dempster NM (2016) Cytotoxicity
of the roots of Trillium govanianum against breast (MCF7), liver (HepG2), lung (A549) and
urinary bladder (EJ138) carcinoma cells. Phytother Res 30:1716–1720
68. Okigbo RN, Anuagasi CL, Amadi JE (2009) Advance in selected medicinal and aromatic
plants indigenous to Africa. J Med Plants Res 3:86–95
69. Li Y, Liu C, Xiao D, Han J, Yue Z, Sun Y, Fan L, Zhang F, Meng J, Zhang R, Wang Z (2015)
Trillium tschonoskii steroidal saponins suppress the growth of colorectal cancer cells in vitro
and in vivo. J Ethnopharmacol 168:136–145
70. Kundra R, Samant S, Sharma RK (2019) Assessment of antioxidant potential of Trillium
govanianum Wall ex. D. Don, a critically endangered medicinal plant of Northestern Indian
Himalaya. Proc Natl Acad Sci, India. Sect B Biol Sci 90:95–101
71. Raghuram TC, Sharma RD, Sivakumar B, Sahay BK (1994) Effect of fenugreek seeds on intra-
venous glucose disposition in non-insulin dependent diabetic patients. Phytother Res 8:83–96
72. Chauhan NS (1999) Medicinal and aromatic plants of Himachal Pradesh. Indus Publishing,
New Delhi
73. Sagar A, Thakur L, Thakur JS (2017) Studies on entophytes and antibacterial activity of
Trillium govanianum Wall. ex D. Don. Int J Bot Stud 2:63–67
74. Zhu L, Tan J, Wang B, Guan L, Liu Y, Zheng C (2011) In-vitro antitumor activity and antifun-
gal activity of Pennogenin steroidal saponins from Paris polyphylla var. yunnanensis, Iran. J
Pharm Res 10:279–286
75. Sharma S, Sharma A, Mehta V, Chauhan RS, Malairaman U, Sood H (2016) Efficient hydroal-
coholic extraction for highest diosgenin content from trillium govanianum (nag chhatri) and
it’s in vitro anticancerous activity. Asian J Pharm Clin Res 9:386–392
76. Sharma S, Mehta V, Sharma P, Jaggi K, Udayabanu M, Sood H (2018) Antifertility activity
and contraceptive potential of the hydro alcoholic rhizome extract of Trillium govanianum in
female wistar rats. Asian J Pharm Clin Res 11:329–332
77. Pawliczak R (2003) The role of radical oxygen species in airway inflammation. Pol Merkur
Lekarski 14:493–496
78. Salvemini D, Wang ZQ, Bourdon DM, Stern MK, Currie MG, Manning PT (1996) Evidence
of peroxynitrite involvement in the carrageenan-induced rat paw edema. Eur J Pharmacol
303:217–220
79. Fridovich I (1997) Superoxide anion radical (O2), superoxide dismutases, and related matters.
J Biol Chem 272:18515–18517
80. Cuzzocrea S, Mazzon E, Dugo L, Serraino I, Ciccolo A, Centorrino T, De Sarro A, Caputi AP
(2001) Protective effects of n-acetylcysteine on lung injury and red blood cell modification
induced by carrageenan in the rat. FASEB J 15:1187–1200
408 K. Rashid et al.
81. Ur Rahman S, Adhikari A, Ismail M, Shah MR, Khurram M, Shahid M, Ali F, Haseeb A,
Akbar F, Iriti M (2016) Beneficial effects of Trillium govanianum rhizomes in pain and inflam-
mation. Molecules 8:20–21
82. Saez TG, Oliva MR, Muniz P, Valls V, Iradi A, Ramos M (1994) Oxidative stress and genetic
damage. In: Health and orange. Fundacion Valenciana de Estudios Avanzados, Valencia,
Spain, pp 51–60
83. Maxwell SR (1995) Prospects for the use of antioxidant therapies. Drugs 49:345–361
84. Braca A, Sortino C, Politi M, Morelli I, Mendez J (2002) Antioxidant activity of flavonoids
from Licania licaniaeflora. J Ethnopharmacol 79:379–381
85. Dasgupta A, Klein K (2014) Antioxidants in food, vitamins and supplements. Prevention and
treatment of disease. Elsevier, pp 277–294
86. Yazdanparast R, Ardestani A (2007) In vitro antioxidant and free radical scavenging activity of
Cyperus rotundus. J Med Food 10:667–674
87. Saleem M, Afza N, Anwar MA, Hai SMA, Ali MS (2003b) A comparative study of essential
oil of C. citratus and some members of the genus citrus. Nat Prod Res 17:369–373
88. Kim DH, Shim SB, Kim NJ, Jang IS (1999) Beta-glucuronidase-inhibitory activity andhepato-
protective effect of Ganoderma lucidum. Biol Pharm Bull 22:162–164
89. Saleem M, Afza N, Anwar MA, Hai SMA, Ali MS, Shujrat S, Rahman A (2003a) Chemistry and
biological significance of essential oils C. Citratus from Pakistan. Nat Prod Res 17:159–163
Comprehensive Review on Wild Basil
Genus Orthosiphon of Lamiaceae
1 Introduction
Phytochemicals possess significant biological activity and are used in human ther-
apy, agriculture, and scientific research [1]. Scientific validation of such phyto-
chemical constituents has been used to discover novel therapeutic agents and
synthesize complex chemical substances [2]. Molecular biology has become tools
and assays prerequisite factors for medicinal plant drug discovery by determining
and implementing appropriate screening assays directed toward physiologically rel-
evant molecular targets. Modern pharmacognosy also encapsulates all these rele-
vant novel arenas of a distinct interdisciplinary science [3].
For cultivating and processing medicinal plants and for manufacturing herbal
medicines, agro-industrial technologies must be applied [4]. The importance of tra-
ditional medicine has been well recognized by the World Health Organization
(WHO) which has created strategies, guidelines, and standard procedures for botan-
ical medicines. Medicinal plants are the resources of novel drug sources, and many
modern treatments are produced indirectly from them. The development and recog-
nition of medicinal plants increase at an exponential rate in both industrialized and
developing nations [5]. The manufacture of morphine on an industrial scale by
E. Merck, Germany, in 1826 marks the beginning of the commercialization of plant-
derived drugs [6]. Nearly half of the top-selling pharmaceuticals in 1991 were based
on either natural products or their analogs derived from them [7].
Flowering plants embark on the most numerous, highly diverse, and successful
extant higher plants in the tropics. India is one of the megadiversity nations with
wide variations in climate, soil, altitude, latitude, and all known types of
agroclimatic, ecologic, and edaphic conditions [8]. The present knowledge records
over 47,500 species of plants belonging to various groups documented from India
[9]. Angiosperms from the most dominant and conspicuous vegetation cover over
18,000 species, representing more than 11.4% of the world’s known flowering plant
species. It has about 19,530 flowering plants, of which 5400 are endemic [10]. On a
global basis, the International Union of Conservation of Natural (IUCN) has esti-
mated that about 12.5% of the world’s vascular plants, totaling about 34,000 spe-
cies, are under varying degrees of extinction. The International Union of
Conservation of Natural resources (IUCN) has enlisted 560 Red List of threatened
plant species in India, of which 247 species are on the red list of threatened species
category [11].
Drug research based on ethnobotanical evidence discovered hundreds of benefi-
cial compounds and pharmacologically active substances in nature, including com-
mon drugs such as aspirin, digoxin, quinine, and opium. Natural constituents have
become the basis and great promise for novel drug sources, and it gratifies abiding
interest in plant-derived medicines described in the folkloric claims of various
countries [12]. In the Indian traditional system of medicine, plants attain novel
interest in finding novel lead molecules for treating various ailments. Medicinal
plants contain substances used for therapeutic purposes or as precursors to synthe-
size valuable drugs due to their inherent active ingredients [13]. Herbal products are
produced by extraction, fractionation, purification, concentration, or other physical
or biological processes of plant materials [14].
In the last few decades, global interest in studying various medicinal plants has
rapidly increased. Investigations have focused on medicinal plants as potential
regimes for treating oxidant-induced diseases. The plant-based chemical constitu-
ents could protect against chronic oxidative stress-related diseases because they
contain variable contents of chemical families and good antioxidants. Certain
classes of secondary metabolites namely, vitamins, and carotenoids have been wit-
nessed in several experiments suggesting that natural compounds have more robust
inhibitive ability than synthetic radical inhibitors. They are natural agents character-
ized by numerous biological activities used in humans, such as antimicrobial, anti-
fungal, anti-inflammatory, antiproliferative, and countless other physical activities
[15]. However, drug development from natural resources is also associated with
certain disadvantages; particularly, it is expected that about 25,000 plant species
would cease to exist by the end of this century [16]. So, it is necessary to identify
novel drug lead molecules for the treatment from alternate sources with low prices,
high efficiency, and fewer side effects. Hence, revealing new indigenous medicinal
herbs is the required uphill task for this recent scenario.
Hence, the current study was focused on scientifically validating the traditional
importance, phytochemical constituents, and biological activity of the endemic
plant species, Orthosiphon. Based on the varied reasons, assessments and explora-
tion of its potential are essential for conserving them from extinction. In this point
of view, in the present study, detailed investigation on the validation of phytochemi-
cal constituents and their role on metabolism have been majorly emphasized to
Comprehensive Review on Wild Basil Genus Orthosiphon of Lamiaceae 411
3 Phytochemical Constituents
Since ancient times, herbs and spices have improved food‘s sensory characteristics,
nutritional, and health properties. Herbs and spices are generally recognized as safe
(GRAS) and excellent chemical additive substitutes. Essential oils are mixtures of
volatile compounds obtained mainly by steam distillation from medicinal and aro-
matic plants, and they are an alternative source to synthetic additives for the food
industry. They are considered potential sources of natural food preservatives due to
the growing interest in developing safe, effective raw food and additives in order to
reduce food demand. Lamiaceae is one of the most influential families producing
essential oils with potent antioxidant and antimicrobial properties. Aromatic plants
are rich in essential oils and are mainly found in the Mediterranean region, where
such oils are a good source of ecological and economic development. The use of
essential oils with antimicrobial and antioxidant properties to increase the shelf life
of food is a promising technology. The vital oils of the family Lamiaceae such as
rosemary, thyme, and sage have been studied extensively concerning their use as
food preservatives [24].
The phytochemical research approach is considered effective in discovering the
bioactive profile of plants of therapeutic importance. Phytochemicals are essential
in cosmetic and medicinal preparations as antimicrobial agents and antioxidants.
The application of investigated plant species in various medicinal aspects is based
on their phytochemical constituents and pharmacological activities. Herbal-based
medicines have gained more popularity due to less toxicity and easy accessibility,
along with technological advances in manufacturing processes [25].
Therefore, it is imperative to search for novel, practical, accessible drug sources
without toxicity. There has been a broad spectrum of action to face various mala-
dies, and medicinal plants are considered as a significant asset [26]. Hyptis suaveo-
lens and Ocimum gratissimum plants were endowed with nutrients, mineral
compounds, and secondary metabolites (flavonoids, alkaloids, tannins, phenolic
compounds, saponins, steroids, glycosides, and essential oils). Antibacterial, anti-
fungal, antioxidant, antiparasitic, antidiabetic, anticancer, antiulcer, wound healing,
and insecticidal activities were well reported for both the species, H. suaveolens and
O. gratissimum. Further, in phytochemical profiling, both the plant species also evi-
denced substantial variation in essential oil composition. This variation is the
414 K. Abirami et al.
consequence of several chemotypes of essential oils, which can influence the bio-
logical activities of the species [27].
Phytochemical screening also revealed that sterols, terpenoids, tannins, sapo-
nins, alkaloids, flavonoids, and glycosides contributed to the medicinal aspects of
the Lamiaceae. This research has provided insight into the use of secondary metab-
olites in traditional medicine in maintaining proper human health [23]. O. aristatus
(Blume) Miq. of the family Lamiaceae is called kumis kucing in Indonesia, which
is a valuable medicinal plant known for its pharmacological properties. Fifteen gen-
otypes of O. aristatus obtained from the ethanolic leaf extracts were undertaken
based on its phytochemical content and pharmacological activities. Chemometric
analysis was also used to investigate the genetic variability based on the phyto-
chemical content and pharmacological activities of O. aristatus genotypes [28].
Extensive research has been carried out on O. stamineus Benth. (Lamiaceae)
since 1930s. Phytochemical studies reported 116 isolated compounds from O. sta-
mineus. Different in vitro and in vivo model studies have been addressed, along with
a survey of all phytochemicals identified in this plant O. stamineus, including flavo-
noids, terpenoids, and essential oils. Previous studies revealed that O. stamineus
possess several pharmacological activities attributed to its phytochemical content. It
has broad conventional and pharmacological uses in various pathophysiological
conditions [29]. Phytochemical studies for O. stamineus also revealed the presence
of abundant bioactive compounds, including terpenoid, phenolic compounds, flavo-
noids, saponin, essential oil, and organic acids [30]. Choo et al. [31] also registered
that the ethanolic extract of O. stamineus, exhibited anticonvulsive activity in
Zebrafish Choo, Kundap [32]. Coelho et al. [33] demonstrated the anticonvulsant
potential of rosmarinic acid in mice, which is considered as an active chemical con-
stituent in the extract Coelho, Vieira [34]. The proteins extracted from the leaves of
O. stamineus leaves may also hold valuable protective potential for central nervous
system (CNS) disorders such as epilepsy.
3.1 Essential Oil
composition of each essential oil. Essential oils with high thymol and carvacrol
determined the most potent antimicrobial activity. As expected, these two com-
pounds demonstrated an interesting antifungal efficacy against the filamentous fun-
gus T. rubrum. Results confirmed that some of the species of Lamiaceae members
used in ethnopharmacological practices as antimicrobial agents possess antibacte-
rial and antifungal potential consistent with their use in alternative or complemen-
tary medicine [35].
was analyzed for GC-FID/MS. The main components of the volatile organic com-
ponents were p-cymene (25.5%), eucalyptol (9.8%), limonene (5.0%), sabinene
(4.8%), carvacrol (3.7%), E-linalool oxide (3.3%), Z-linalool oxide (3.0%) in
N. conferta; carvacrol (47.3%), p-cymene (15.8%), γ-terpinene (8.6%), myrcene
(8.6%), caryophyllene (2.0%) in O. onites and carvacrol (32.6%), p-cymene
(22.2%), γ-terpinene (15.1%), myrcene (5.5%), and caryophyllene (3.3%) in
S. cuneifolia. Carvacrol was the most abundant component in the volatile organic
compounds of O. onites (47.3%), S. cuneifolia (32.6%), and N. conferta (3.7%) [45].
The hydro-distilled essential oil from the aerial parts of O. pallidus Royle, ex
Benth (Lamiaceae), was investigated using gas chromatography equipped with
flame ionization detector (GC-FID) and gas chromatography coupled with mass
spectrometry (GC-MS). Fifty-two compounds representing 98.4% of the total oil
constituents were identified. The major constituents were β-caryophyllene (17.4%)
and 7-epi-α-selinene (15.2%). The other minor constituents were terpinolene
(6.9%), β-pinene (6.8%), β-element (5.1%), α-humulene (4.9%), α-copaene (4.8%),
epi-cubebol (4.5%), and zonarene (3.9%). The oil was found to be rich in sesquiter-
pene hydrocarbon-type constituents [46].
The chemical composition of the essential oil of O. diffusus was investigated.
Extraction by hydrodistillation followed by gas chromatography and mass spec-
trometry (GC-MS) yielded 25 compounds representing 95.3% of the oil. The major
volatile components of the oil were n-eicosane (19.5%), t-caryophyllene (18.6%),
octocosane (12.2%), limonene (11.6%), β-ocimene (4.2), methyl palmitate (2.8%),
and elemol (2.6%) [47].
The chemical composition of the volatile oil from the leaves of Endostemon
obtusifolius (E.Mey. ex Benth.) N.E.Br. was investigated. The composition of oils
obtained by hydrodistillation followed by gas chromatography and mass spectrom-
etry (GC-MS) yielded 50 compounds representing 99.8% of the oil. The major vola-
tile components of the oil were phenol (26.92%), 1,3,6,10-cyclotetradecatetraene,
3,7,11-trimethyl-14-(1-methylethyl)-,[S-(E,Z,E,E)] (19.13%), acetic acid,
1,7,7-trimethyl-bicyclo[2.2.1]hept-2-yl ester (6.44%), cyclooctene (5.25%),
1H-cyclopropa[a]naphthalene,decahydro-1,1,3a-trimethyl-7-methylene-[1aS-(1a.
alpha.,3a.alpha.,7a.beta.,7b.alpha.)] (4.98%), 3-cyclohexen-1-ol,4-methyl-1-(1-
methylethyl) (3.81%), cycloisolongifolene, 8,9-dehydro (3.52%), and
1H-cycloprop[e] azulene, decahydro-1,1,7-trimethyl-4-methylene (3.12%). Phenol
was the major compound detected from E. obtusifolius used as an oral anesthetic
and analgesic agent to treat pharyngitis. It is considered as a versatile precursor for
large collection of drugs including aspirin, herbicides, and pharmaceutical
drugs [47].
second source of diversity is due to the layering of functional groups and substitu-
ents upon the structural scaffolds in distinct regio- and stereospecific manners.
Sesquiterpenes also include essential oils and aromatic constituents with several
pharmacological activities [49, 50].
Essential oils are widely used in pharmaceutical, sanitary, cosmetic, agriculture,
and food industries for their bactericidal, viricidal, fungicidal, antiparasitical, and
insecticidal properties. Their anticancer activities are well documented. Over a hun-
dred essential oils from more than 20 plant families have been tested on more than
20 types of cancers in last 10 years [48]. In addition, cytotoxic effects have been
reported for many essential oils [51, 52]. However, very few studies have been done
on the combination of essential oils and their major compounds to find assumed
synergistic constructive effects.
In vitro and in vivo studies have shown that sinensetin possesses strong anticancer
activities and a wide range of pharmacological activities such as anti-inflammatory,
antioxidant, antimicrobial, antiobesity, antidementia, and vasorelaxant activities
[55]. Eupatorin, as a major bioactive flavonoid constituent in O. stamineus, pos-
sesses numerous strong biological activities, including anticancer, anti-inflammatory,
and vasorelaxation activities [53, 56–62].
Plant-derived compounds with high therapeutic effects are gaining more concern
all over the Globe. Researchers are mainly targeting ethnic and folkloric knowledge
to explore novel drug candidates. Orthosiphon thymiflorus is a medicinal, aromatic
herb used in folkloric medicine and in Ayurvedic preparations. Hence, it is impera-
tive to evaluate the cytotoxic potential and chemical profile of the plant [63].
Phytochemical profiling of O. diffusus has resulted in the isolation of four relatively
rare, novel polychiral furanopyrans, namely, orthodiffenes A–D, characterized from
the detailed studies of their 1D and 2D NMR spectral analysis. The X-ray crystal-
lographic analysis of orthodiffene A was also accomplished. The in vitro cytotoxic
activity of orthodiffenes A–C was tested against Jurkat and HL-60 cells using camp-
tothecin as a positive control. Orthodiffenes A and B showed comparable camptoth-
ecin activity against HL-60 and Jurkat cells, respectively [64].
Due to its unique ability to bind with CB2 receptors, beta-caryophyllene has potent
anti-inflammatory, antimicrobial, antibacterial, and antioxidant properties. It is
known to relieve anxiety, pain, reduce cholesterol, prevent osteoporosis, and treat
seizures. Also, some research has shown that it may help against certain neurode-
generative diseases and cancers. It is considered as a therapeutic agent to prevent
and treat osteoporosis [65]; it acts as a local anesthetic agent by releasing the same
endorphins as morphine without any additives. Additionally, when used together
420 K. Abirami et al.
The publication trend shows that increasing interest in medicinal plant research and
analysis is reflected in the number of recent publications, with more than a threefold
increase from 4686 publications during the year 2008 to 14,884 in 2018. Output
published during the 8 years of the present decade alone outnumbered all those
combined before 2000 [66].
There is good scope for medicinal plants as there are about half a million plants
worldwide, and most of them were not investigated for their medicinal properties.
The hidden potential of therapeutic activities could be decisive in treating diseases
[67]. Among the variety of modern medicines, many are produced indirectly from
medicinal plants, such as aspirin. Studying medicinal plants helps to understand
plant toxicity and protects humans and animals from synthetic poison. The medici-
nal effects of plants are due to the nature of secondary metabolites inherent in them.
Keeping this in consideration, there has been a renowned interest in the field of
natural product chemistry. This interest can be due to several factors, including ther-
apeutic needs, the remarkable diversity of both chemical structure and biological
activities of naturally occurring secondary metabolites, the utility of novel bioactive
natural compounds as biochemical probes, the development of novel and sensitive
techniques to detect biologically active natural products, improved approaches to
isolate, purify, and structurally characterize these bioactive constituents and
advances in solving the demand for supply of complex natural products [68].
Most of the pharmaceutical industry is highly dependent on the wild population
to supply raw materials to extract medicinally essential compounds. Unfortunately,
the genetic diversity of medicinal plants in the world is getting endangered at an
alarming rate due to ruinous harvesting practices and over-harvesting for the pro-
duction of medicines, with little or no regard for the future. Also, extensive destruc-
tion of plant-rich habitats has been made due to forest degradation, agriculture
encroachment, and urbanization. In modern medicine, plants are used as a source of
direct therapeutic agents, a model for new synthetic compounds, and a taxonomic
marker for elaborating more complex semi-synthetic chemical compounds [69].
The ability to process data using multivariate analysis software has opened up
new vistas to metabolomics, giving us greater capacity to understand many
Comprehensive Review on Wild Basil Genus Orthosiphon of Lamiaceae 421
References
1. Vasu K, Goud JV, Suryam A, Singara MA (2009) Biomolecular and phytochemical analyses
of three aquatic angiosperms. Afr J Microbiol Res 3(8):418–421
2. Akrout EI, Jani H, Zammouri T, Mighri H, Neffati M (2010) Phytochemical screening and
mineral contents of annual plants growing wild in the southern of Tunisia. J Phytology
2(1):034–040
3. Sarker SD (2012) Pharmacognosy in modern pharmacy curricula. Pharmacogn Mag 8(30):91
422 K. Abirami et al.
4. World Health Organization (1993) Research guidelines for evaluating the safety and efficacy
of herbal medicines. WHO Regional Office for the Western Pacific, Manila
5. World Health Organization (1998) Regulatory situation of herbal medicines. A worldwide
review, Geneva, pp 1–5
6. Galbley S, Thiericke R (1999) Drug discovery from nature, Series: Springer Desktop Editions
in Chemistry
7. Cragg GM, Newman DJ (2013) Natural products: a continuing source of novel drug leads.
Biochim Biophys Acta 1830(6):3670–3695
8. Dar JA, Subashree K, Sundarapandian S et al (2019) Invasive species and their impact on
tropical forests of Central India: a review. In: Tropical ecosystems: Structure, functions and
challenges in the face of global change, pp 69–109
9. Raju VS, Reddy CS, Suthari S (2010) Flowering plant diversity and endemism in India: an
overview. Algae 2(40,000):6–25
10. Singh R (2015) Medicinal plants: a review. J Plant Sci 3(1):50–55
11. Rajasekharan PE, Wani SH (2020) Conservation and utilization of threatened medicinal
plants. Springer Nature- Science, p 565
12. Rangari VD (2007) Pharmacognosy & Phytochemistry, 1st edn. Career Publication,
Pune, pp 4–7
13. Okigbo RN, Eme UE, Ogbogu S (2008) Biodiversity and conservation of medicinal and
aromatic plants in Africa. Biotechnol Mol Biol Rev 3(6):127–134
14. Kumar P, Mishra S, Malik A, Satya S (2011) Insecticidal properties of Mentha species: a
review. Ind Crop Prod 34(1):802–817
15. Lourenco SC, Moldao-Martins M, Alves VD (2019) Antioxidants of natural plant origins:
from sources to food industry applications. Molecules 24(22):4132
16. Mahidol C, Ruchirawat S, Prawat H et al (1998) Biodiversity and natural product drug dis-
covery. Pure Appl Chem 70(11):2065–2072
17. Sundarammal S, Thirugnanasampandan R, Selvi MT (2012) Chemical composition analy-
sis and antioxidant activity evaluation of essential oil from Orthosiphon thymiflorus (Roth)
Sleesen. Asian Pac J Trop Biomed 2:S112–S115
18. Mishra LK, Sarkar D, Shetty K (2019) Human health-relevant bioactives and associated
functionalities of herbs in the Lamiaceae family. In: Functional Foods and Biotechnology.
CRC Press, pp 115–131
19. Chithra V, Adersh M, Reji SR, Nair GM (2013) Screening biological activities of Orthosiphon
aristatus. Int J Adv Res 5:594–600
20. Heinrich M, Gibbons S (2001) Ethnopharmacology in drug discovery: an analysis of its role
and potential contribution. J Pharm Pharmacol 53(4):425–432
21. Ghaffari H, Venkataramana M, Nayaka SC et al (2013) Hepatoprotective action of
Orthosiphon diffusus (Benth.) methanol active fraction through antioxidant mechanisms: an
in vivo and in vitro evaluation. J Ethnopharmacol 149(3):737–744
22. Adnyana IK, Setiawan F, Insanu M (2013) From ethnopharmacology to clinical study of
Orthosiphon stamineus Benth. Studies 1(2)
23. Okach DO, Nyunja ARO, Opande G (2013) Phytochemical screening of some wild plants
from Lamiaceae and their role in traditional medicine in Uriri District-Kenya. Int J Herb Med
1(5):135–143
24. Nieto G (2017) Biological activities of three essential oils of the Lamiaceae family.
Medicines 4(3):63
25. Masih NG, Singh BS (2012) Phytochemical screening of some plants used in herbal based
cosmetic preparations. In: Chemistry of Phytopotentials: health, energy and environmental
perspectives. Springer, Berlin, Heidelberg, pp 111–112
26. Garba ABH, Arya MA, Traore A, Ouedraogo S (2017) Etude des effets vermicide et anti-
diarrheique du macere aqueux des feuilles de Salvadora persica, L. (Salvadoraceae). Int J
Biol Chem 11(1):54–66
Comprehensive Review on Wild Basil Genus Orthosiphon of Lamiaceae 423
27. Degla LH, Olounlade PA, Amoussa AMO et al (2021) Pharmacological and biochemical
aspects of the Lamiaceae Family used in the treatment of intestinal Parasitosis in West and
Central Africa. Pharmacogn Rev 15(29):69–75
28. Batubara I, Komariah K, Sandrawati A et al (2020) Genotype selection for phytochemical
content and pharmacological activities in ethanol extracts of fifteen types of Orthosiphon
aristatus (Blume) Miq. leaves using chemometric analysis. Sci Rep 10:20945
29. Ameer OZ, Salman IM, Asmawi MZ, Ibraheem ZO (2012) Orthosiphon stamineus: tra-
ditional uses, phytochemistry, pharmacology, and toxicology. Mun Fei Yam J Med Food
15:8678–8690
30. Maheswari C, Venkatnarayanan R, Manavalan R et al (2015) Phytochemical screening and
in vitro free radical scavenging activity of Orthosiphon stamineus and Coccinia grandis. Int
Res J Pharm 6:627–630
31. Choo BKM, Kundap UP, Kumari Y, Hue SM et al (2018) Orthosiphon stamineus leaf extract
affects TNF-and seizures in a zebrafish model. Front Pharmacol 9:139
32. Zhu HL, Wan JB, Wang YT et al (2014) Medicinal compounds with antiepileptic/anticonvul-
sant activities. Epilepsia 55:3–16
33. Coelho VR, Vieira CG, De Souza LP et al (2015) Antiepileptogenic, antioxidant and geno-
toxic evaluation of rosmarinic acid and its metabolite caeic acid in mice. Life Sci 122:65–71
34. Rabiei Z (2017) Anticonvulsant effects of medicinal plants with emphasis on mechanisms of
action. Asian Pac J Trop Biomed 7:166–172
35. Khoury M, Stein D, Eparvier V et al (2016) Report on the medicinal use of eleven Lamiaceae
species in Lebanon and rationalization of their antimicrobial potential by examination of the
chemical composition and antimicrobial activity of their essential oils. eCAM 2016:17
36. Memar Mohammad Y, Parisa R, Naser A et al (2017) Carvacrol and thymol: strong antimicro-
bial agents against resistant isolates. Rev Med Microbiol 28(2):63–68
37. Fizur NMM, Hayate J, Hasan AT et al (2017) Pharmacological properties and molecular
mechanisms of thymol: prospects for its therapeutic potential and pharmaceutical develop-
ment. Front Pharmacol 8:380
38. Lee KW, Everts H, Kappert HJ et al (2003) Dietary Carvacrol lowers body weight gain but
improves feed conversion in female broiler chickens. J Appl Poult Res 12(4):2003
39. Namata Abba B, Romane A, Ilagouma AT (2020) Antibacterial activity of Endostemon tereti-
caulis (Poir.) M. Ashby essential oil and ethanolic extract against resistant pathogenic bacte-
ria. Nat Prod Commun 15(9):1–9
40. Robertson DG (2005) Metabonomics in toxicology: a review. Toxicol Sci 85:809–822
41. Kanthal LK, Dey A, Satyavathi K, Bhojaraju P (2014) GC-MS analysis of bio-active com-
pounds in methanolic extract of Lactuca runcinata DC. Pharm Res 6(1):58
42. Pakkirisamy M, Kalakandan SK, Ravichandran K (2017) Phytochemical screening, GC-MS,
FT-IR analysis of Methanolic extract of Curcuma caesia Roxb (Black Turmeric). Pharm J
9(6):952–956
43. Hajdari A, Mustafa B, Hyseni L et al (2020) Phytochemical study of eight medicinal plants of
the lamiaceae family traditionally used as tea in the Sharri Mountains region of the Balkans.
Sci World J 2020:9
44. Asmira AREN, Azlini I, Nor OM et al (2018) GC-MS analysis of phytochemical com-
pounds in Syzygium polyanthum leaves extracted using ultrasound-assisted method. Pharm
J 10(1):110–119
45. Yayli B, Tosun G, Karaköse M et al (2014) SPME/GC-MS analysis of volatile organic com-
pounds from three Lamiaceae species (Nepeta conferta Hedge & Lamond, Origanum onites
L. and Satureja cuneifolia Ten.) growing in Turkey. Asian J Chem 26(9):2541
46. Joshi RK (2020) GC-MS analysis of the volatile constituents of Orthosiphon pallidus Royle,
ex Benth. Nat Prod Res 34(3):441–444
47. Sadashiva CT, Naidoo Y, Naidoo J R, Naidoo G (2013) Chemical composition of the essen-
tial oil from the leaves of Endostemon obtusifolius (E.Mey. ex Benth.) N.E.Br. Biochem
Pharmacol 2:4
424 K. Abirami et al.
48. Bayala B, Bassole IHN, Scifo R et al (2014) Anticancer activity of essential oils and their
chemical components - a review. Am J Cancer Res 4(6):591–607
49. Chappell J, Robert M (2010) Coates, 1.16 - Sesquiterpenes. In: (Ben) Liu H-W, Mander L
(eds) Comprehensive Natural Products II. Elsevier, pp 609–641
50. Awouafack MD, Tane P, Kuete V, Jacobus N (2013) Eloff, 2 - Sesquiterpenes from the medici-
nal plants of Africa. In: Kuete V (ed) Medicinal Plant Research in Africa. Elsevier, pp 33–103
51. Cavalieri E, Mariotto S, Fabrizi C et al (2004) Alpha-Bisabolol, a nontoxic natural compound,
strongly induces apoptosis in glioma cells. Biochem Biophys Res Commun 315:589–594
52. Lampronti I, Saab AM, Gambari R (2006) Antiproliferative activity of essential oils derived
from plants belonging to the Magnoliophyta division. Int J Oncol 29:989–995
53. Yam MF, Lim V, Salman IM et al (2010) HPLC and anti-inflammatory studies of the flavonoid
rich chloroform extract fraction of Orthosiphon stamineus leaves. Molecules 15:4452–4466
54. Akowuah GA, Ismail Z, Norhayati I et al (2005) The effects of different extraction solvents
of varying polarities on polyphenols of Orthosiphon stamineus and evaluation of the free
radical-scavenging activity. Food Chem 93:311–317
55. Han Jie L, Jantan I, Yusoff SD et al (2021) Sinensetin: an insight on its pharmacological
activities, mechanisms of action and toxicity. Front Pharmacol 11:553404
56. Razak NA, Abu N, Ho WY et al (2019) Cytotoxicity of eupatorin in MCF-7 and MDA-MB-231
human breast cancer cells via cell cycle arrest, anti-angiogenesis and induction of apoptosis.
Sci Rep 9:1514
57. Lee K, Hyun Lee D, Jung YJ et al (2016) The natural flavone eupatorin induces cell cycle
arrest at the G2/M phase and apoptosis in HeLa cells. Appl Biol Chem 59:193–199
58. Estevez S, Marrero MT, Quintana J (2014) Eupatorin-induced cell death in human leukemia
cells is dependent on caspases and activates the mitogen-activated protein kinase pathway.
PLoS One 9:e112536
59. Androutsopoulos V, Arroo RRJ, Hall JF et al (2008) Antiproliferative and cytostatic effects
of the natural product eupatorin on MDA-MB-468 human breast cancer cells due to CYP1-
mediated metabolism. Breast Cancer Res 10(3):R39
60. Doleckova I, Rarova L, Gruz J et al (2012) Antiproliferative and antiangiogenic effects of fla-
vone eupatorin, an active constituent of chloroform extract of Orthosiphon stamineus leaves.
Fitoterapia 83:1000–1007
61. Laavola M, Nieminen R, Yam MF et al (2012) Flavonoids eupatorin and sinensetin present in
Orthosiphon stamineus leaves inhibit inflammatory gene expression and STAT1 activation.
Planta Med 78:779–786
62. Yam MF, Tan CS, Ahmad M, Shibao R (2016) Mechanism of vasorelaxation induced by
eupatorin in the rats aortic ring. Eur J Pharmacol 789:27–36
63. Devi SR, Thoppil JE (2016) Cytotoxic studies and phytochemical analysis of Orthosiphon
thymiflorus (Roth) Sleesen. Int J Pharm Sci 8(2):249–255
64. Holla H, Srinivas Y, Majhi A et al (2011) Novel cytotoxic constituents of Orthosiphon dif-
fusus. Tetrahedron Lett 52(1):49–52
65. Yamaguchi M, Levy RM (2016) β-Caryophyllene promotes osteoblastic mineralization, and
suppresses osteoclastogenesis and adipogenesis in mouse bone marrow cultures in vitro. Exp
Ther Med 12(6):3602–3606
66. Martin F, Michael H, Anthony B (2020) Medicinal plant analysis: a historical and regional
discussion of emergent complex techniques. Front Pharmacol 10
67. Singh R (2015) Medicinal plants: a review. J Plant Sci, Special Issue: Medicinal Plants
3(1–1):50–55
68. Clark AM (1996) Natural products as a resource for new drugs. Pharm Res 13(8):1133–1141
69. Karthika C, Manivannan S (2018) Pharmacognostic, physicochemical analysis and phyto-
chemical screening of the leaves of W. trilobata. L. Int J ChemTech Res 11(02):124–131
70. Chen SL, Yu H, Luo HM et al (2016) Conservation and sustainable use of medicinal plants:
problems, progress, and prospects. Chin Med 11:37
Comprehensive Review on Wild Basil Genus Orthosiphon of Lamiaceae 425
92. Yuniarto A, Susilawati ELIS, Khairunnisa ISMI et al (2017) Antioxidant and gastric ulcer
healing effect of Orthosiphon stamineus (Benth.) leaves extract in aspirin-induced rats. Asian
J pharm. Clin Res 10(2):397–399
93. Movahedi A, Basir R, Rahmat A et al (2015) Orthosiphon stamineus: an Asian tea with sub-
stantial anticancer properties. J Nutr Sci Diet:44–52
94. Nair GM (2011) Evaluation of antioxidant properties of some species of Lamiaceae. J Med
Aromat Plant Sci 33(1):27–30
95. Alshawsh MA, Abdulla MA, Ismail S et al (2011) Hepatoprotective effects of Orthosiphon
stamineus extract on thioacetamide-induced liver cirrhosis in rats. Evidence-Based
Complement Alternat Med 2011:103039
96. Yam MF, Basir R, Asmawi MZ et al (2007) Antioxidant and hepatoprotective effects of
Orthosiphon stamineus benth. Standardized extract. Am J Chin Med 35:115–126
97. Yam MF, Asmawi MZ, Basir R (2008) An investigation of the anti-inflammatory and analge-
sic effects of Orthosiphon stamineus leaf extract. J Med Food 11:362–368
98. Kavimani S, Ilango R, Thangadurai JG et al (1997) Diuretic activity of aqueous extract of
Orthosiphon thymiflorus in rats. Indian J Pharm Sci 59(2):96
99. Sini KR, Haribabu Y, Sajith MS, Sreekumar SK (2012) In-vitro Cytotoxic activity of
Orthosiphon thymiflorus(Roth.) sleensen leaf extract against dalton lymphoma ascites cell
line. J Chem Pharm Res 4(1):917–921
100. Mercy Lavanya S, Gnanamani A, Ilavarasan R (2015) Evaluation of the antibacterial activity
of the extracts of the whole plant of Orthosiphon thymiflorus (Roth.) Sleesen. J Chem Pharm
Res 7(2):872–875
101. Neilson EH, Goodger JQ, Woodrow IE et al (2013) Plant chemical defense: at what cost?
Trends Plant Sci 18(5):250–258
Zingiberaceae Plants: A Cornucopia
of Promising Chemotherapeuticals
for Cancer Cure
1 Introduction
Cancer, the second leading cause of human death worldwide, behind cardiovascular
disease, might soon rank as the leading cause of death. It is the single most impor-
tant barrier to increasing life expectancy in every country of the world in the 21st
century [1]. Cancer is an abnormal growth of cells caused by multiple changes in
gene expression which leads to dysregulation in balancing cell proliferation and cell
death. As a consequence, a population of cells evolve at the site of origin, capable
of invading into tissues at distant sites, causing significant morbidity and death of
the host, if untreated [2–5]. Cancers are conventionally treated using a combination
of three major modes - surgery, radiation, and chemotherapy. Chemotherapy
involves treatment of cancer with one or more chemicals known to have cytotoxic,
antineoplastic activity. Traditional chemotherapeutic agents, which act by killing
rapidly dividing cancer cells, also harm cells that divide rapidly under normal cir-
cumstances, thereby resulting in side effects such as immunosuppression and muco-
sitis among others. Newer anticancer drugs (targeted chemotherapy) are not
indiscriminately cytotoxic, but rather target proteins that are abnormally expressed
in cancer cells and are essential for their growth [6]. It is believed that anticancer
effects of plants develop by suppressing pathways involved in cancer progression,
DNA repair, increasing body immunity, and inducing antioxidant effects [7–9].
During the last few decades, ethnomedicinal plants have played a significant role
in the development of anticancer drugs with fewer side effects in different conti-
nents of the world. Out of 121 prescription drugs that are being used today for
cancer treatment, 90 are plant-based and 75% of them were discovered from folk-
lore claims. Secondary metabolites from plants, including alkaloids, terpenoids, and
polyphenolic compounds with promising anticancer potential, have been developed
for clinical practice [10, 11]. Classical examples include Vinca alkaloids (vinblas-
tine and vincristine), isolated from Catharanthus roseus G. Don. (Apocynaceae),
paclitaxel and docetaxel - semisynthetic derivatives of Taxanes (diterpenoids) from
the Pacific Yew tree, Taxus brevifolia and T. baccata respectively, irinotecan and
topotecan - semisynthetic derivatives of Camptotheca alkaloids, isolated from
Camptotheca acuminate and Podophyllum lignans from mayapple tree, Podophyllum
peltatum and P. hexandrum [11–13].
Zingiberaceae is one of the largest families of the plant kingdom, distributed
widely throughout the tropics, particularly in Southeast Asia [14, 15]. The family
consists of a large number of economically and medicinally important plants well
known for their use in ethnomedicine. The Zingiberaceae plants contain a number
of volatile and essential oils including terpenoids, phenylpropanoids, flavonoids,
and sesquiterpenes, which have been reported to possess anticancer activity [16–
19]. Hence, these plants are considered to be excellent candidates for development
of novel chemotherapeutics. Various extracts and pure compounds / secondary
metabolites isolated from Curcuma, Zingiber, Kaempferia, Alpinia, Amomum, and
Hedychium genera reportedly possess anticancer activity as evidenced by in vitro
and in vivo studies [20–24]. Few notable examples for anticancer compound include
curcumin/curcuminoids (Curcuma longa), zerumbone (Zingiber zerumbet), gin-
gerol, and shogaol (Zingiber officinale) among many others [25–29]. Even though a
number of review articles were available about various biological activities of dif-
ferent genera of Zingiberaceae family, a comprehensive scrutiny about anticancer
potential of these plants was found lacking. Hence, this chapter aims to overview
anticancer potential of organic solvent extracts and compounds identified therein
belonging to different genera of Zingiberaceae plants.
2 Zingiberaceae
Kingdom: Plantae
Subkingdom: Trachebionta
Superdivision: Spermatophyta
Division: Magnoliophyta
Subdivision: Angiospermae
Class: Monocotyledonae (Liliopsida)
Subclass: Zingiberidae
Order: Zingiberales
Family: Zingiberaceae
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 429
3 Phytochemistry of Zingiberaceae
The tropical and subtropical genus, Alpinia Roxb., with about 230 species, is mainly
distributed in the Indo-Pacific region [15]. The genus is generally called as ‘shell
ginger’ and several species are cultivated as ornamentals. Alpinia species are well
known medicinal herbs with incredible biopharmaceutical potential. The presence
of bioactive substances such as terpenoids, diarylheptanoids, phenylpropanoids,
and flavonoids is key to their therapeutic efficiency [175]. Many in vitro studies car-
ried out in diverse cancer cell lines and in vivo studies with animal models reflect
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 431
Fig. 1 Molecular structures of some phytochemicals from Zingiberaceae plants reported with
anticancer activities
Table 1 List of anticancer bioactive extracts, fractions, and pure compounds of various genera of
Zingiberaceae family
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
Genus Alpinia Roxb.
A. oxyphylla Fruits Hexane and ethyl acetate Antiangiogenic against [74]
fractions zebrafish model,
human umbilical vein
endothelial cells and
tumor cell lines
A. galanga Rhizome 10S-10-Acetoxychavicol Cytotoxicity against [75]
acetate and p-coumaryl alcohol A549, SNU638,
c-O-methyl ether HT1080, HL60 and
HCT116 human cancer
cell lines
A. conchigera Rhizome 10S-10-Acetoxychavicol Apoptotic induction in [76]
acetate MCF-7, HSC-2,
HSC-4, HepG2 and
CaSki
A. officinarum Rhizome Galangin Prevents skin cancer [77]
7-(3,4-Dihydroxyphenyl)-1-(4- Cytotoxicity against [78]
hydroxy-3-methoxyphenyl)-4- HepG2, MCF-7 and
en-3-heptanone SF-268 cancer cell
lines
Diarylheptanoids Induces mitochondrial [79]
apoptosis and S-phase
cell cycle arrest in
neuroblastoma IMR-32
cell line
A. mutica Rhizome Pinostrobin Cytotoxic against KB, [80]
MCF-7 and Caski
cancer cells
A. purpurata Leaves Ethyl acetate extract Cytotoxicity against [81, 82]
OAW42 and HeLa
cells
A. Rhizome Hexane and ethyl acetate Cytotoxic against KB, [83]
pahangensis extracts CaSki and HCT116
cancer cells
A. murdochii Rhizome Hexane and dichloromethane Cytotoxic effect [84]
and extracts against SKOV-3 cells
leaves
A. katsumadai Seeds Rubraine, isorubraine, and Cytotoxicity against [85]
sumadain HepG2, MCF7 and
MDA-MB-435 cancer
cell lines
(continued)
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 433
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
A. zerumbet Flowers Dichloromethane and methanol Antitumor activity [86]
extracts against Ehrlich Ascites
Carcinoma (EAC) cells
in vivo
Flowers 5,6,dehydrokawain Cytotoxicity against
MCF7, HepG2, HEP-2
cancer cell lines
A. nantoensis Rhizome Ethanol extract Inhibit cell migration [87]
and leaf and invasion MCF-7
extracts and MDA-MB-231
breast cancer cell lines
A. scabra Leaves Hexane and chloroform extract Cytotoxic against [88]
and MCF7 and SKOV-3
rhizome cell lines
A. Seeds Diarylheptanoids Antiproliferative [89]
blepharocalyx against HT-1080
(human) and 26-L5
(murine) carcinoma
cell lines
A. pricei Rhizome Ethanolic extract Induce apoptosis [90]
against KB carcinoma
cell lines through
mitochondria-
dependent pathway
Genus Amomum Roxb.
A. subulatum Fruit Cardamonin Induce apoptosis in [91]
HCT116 cancer cell
lines through extrinsic
apoptotic pathway
Seeds Hexane and ethyl acetate Cytotoxicity against [92]
extracts MCF7 and HeLa
cancer cell lines
A. aculeatum Leaves Aculeatin A and B Cytotoxic against [93]
MCF7 breast cancer
cell line
A. kravanh Fruit Ethanol extract Cytotoxicity against [46]
SMMC-7721 cell lines
(continued)
434 T. Soumya et al.
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
A. tsao-ko Fruit Tsaokoarylone Cytotoxic against A549 [94]
and SK-Mel-2 cancer
cell lines
Essential oil Induces apoptosis [95]
against HepG2 cell line
Isotsaokoin, hannokinol, Cytotoxicity against [96]
2,3-dihydro-2-(4′-hydroxy- HepG-2, SMMC-7721,
phenylethyl)-6-[(3″,4″- HeLa and A549 cancer
dihydroxy-5″-methoxy) cell lines
phenyl]-4-pyrone and
4-dihydro-2-(4′-hydroxy-
phenylmethyl)-6-[(3″,4″-
dihydroxy-5″ methoxyphenyl)
methylene]-pyran-3,5-dione
Ethanol extract Antitumor activity [97]
against ovarian cancer
SKOV3 cells with
antiangiogenic activity
in vivo
A. villosum Seeds Polysaccharides Cytotoxic effects [98]
against human
hepatocellular
carcinoma cell lines
Hep G2
A. verum Shoots Essential oils Cytotoxic against [99]
Human prostate
DU145 cancer cell line
A. xanthioides Seeds Monoterpenoids, Cytotoxicity against [100–103]
Sesquiterpenoids, Terpene SK-OV-3, SK-MEL-2,
Glycosides, Amoxantin A A549 and HCT15
(diterpenoid) cancer cell lines
A. maximum Roots Labdane diterpenoids Cytotoxicity against [104]
and MCF-7, SMMC-7721,
Fruits MG-63 and HepG2
cancer cell lines
Genus Kaempferia L.
K. rotunda Rhizome Pinostrobin Antitumor activity [105]
against human breast
cancer, T47D cell line
(in vitro) and xenograft
model (in vivo)
Lectin Induces apoptosis in [106, 107]
Ehrlich ascites
carcinoma cells, SW48
and SW480 colorectal
cancer cell lines
(continued)
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 435
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
K. galanga Rhizome Alcoholic extracts and Antitumor activity [108, 109]
Ethyl-p-methoxycinnamate against Ehrlich ascites
carcinoma cells and
human
cholangiocarcinoma
CL-6 cell line (in vitro)
as well as in vivo
model
Polysaccharides Antitumor activity on [110]
H22 solid tumor (in
vivo)
Ethyl-p-methoxycinnamate Cytotoxicity against [111]
HSC-3 and Ca922 cell
lines
K. parviflora Rhizome Ethanol extract Apoptosis induction in [112]
leukemic HL60 and
U937 cell lines
inhibition of cell [113, 114]
migration and invasion
and induction of
apoptosis in HeLa
(cervical) and SKOV3
(ovarian) cells
Polymethoxyflavones Cytotoxicity against [115]
human cervical (HeLa)
and gastric
adenocarcinoma (AGS)
cell lines
5,7,4-trimethoxyflavone Cytotoxicity against [116]
human
cholangiocarcinoma
HuCCA-1 and
RMCCA-1 cell lines
K. Rhizome Abietene diterpene and Cytotoxicity against [117]
angustifolia kaempfolienol HL-60 and MCF-7
cancer cell lines
K. elegans Rhizome Labdane and clerodane Cytotoxicity against [118]
K. pulchra Rhizome diterpenoids leukemic HL60 cell
line
Genus Curcuma L.
(continued)
436 T. Soumya et al.
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
C. longa Rhizome Various extracts Cytotoxicity against [119–122]
U937, Molt4, A549,
T98G, HeLa,
MDA-MB-231 human
cancer cell lines and
murine melanoma cell
line, B164A5
Curcuminoids / Curcumin Anticancer activity [123–128]
against multiple human
carcinomas including
melanoma, head and
neck, breast, colon,
pancreatic, prostate and
ovarian cancers
C. amada Rhizome Supercritical CO2 extract Cytotoxicity against [129]
human glioblastoma
(U-87MG) cell line
Rhizome Methanol extracts Cytotoxicity against [130]
and human MCF7 and
leaves MDA-MB-231 breast
cancer cell lines
C. aromatica Rhizome Aqueous extract Induces apoptosis and [131]
G2/M arrest in colon
carcinoma cell
lineLS-174-T
Essential oil Antitumor and [132–134]
chemoprevention
against hepatoma in
mice models (in vivo)
Ethanolic extract Antiangiogenic and [135]
proapoptotic activity in
Ehrlich ascites tumor
model (in vivo)
C. caesia Rhizome Methanol extract Antitumor activity on [136]
Ehrlich’s ascites
carcinoma (EAC)
bearing mice.
Antitumor potential [137]
against DEN-induced
hepatocellular
carcinoma
(continued)
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 437
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
C. zedoaria Rhizome Ethanolic extract Antiproliferative and [138]
invasive activities
against human
esophageal squamous
carcinoma TE-8 cells
(in vitro) and suppress
tumor formation in
mice (in vivo)
Hexane and chloroform Cytotoxicity against [139]
extracts ovarian cancer cells
SKOV3
Essential oil Cytotoxic effects on [140]
gastric cancer AGS
cells and induce cell
cycle arrest and
apoptosis
Cytotoxic and induces [141]
apoptosis in non-small
cell lung carcinoma
H1299 cells and
antitumor activity
against H1299
xenograft mice model
(in vivo)
Antiangiogenic activity [142]
both in vitro and in
vivo -suppressing
melanoma growth and
lung metastasis
Isocurcumenol Cytotoxicity against [21]
human cancer KB,
A549, K562 cell lines
and mice DLA
(Daltons Lymphoma
Ascites) cells
(continued)
438 T. Soumya et al.
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
C. Rhizome Xanthorrhizol Inhibit tumor nodules [143]
xanthorrhiza in a spontaneous
mouse lung metastasis
model (in vivo)
Induce apoptosis via [144, 145]
activation of p53-
dependent
mitochondrial pathway
in HCT 116 MCF 7
and MDA-MB-231
cancer cell lines
Induce caspase- [146]
independent apoptosis
in oral squamous cell
carcinoma SCC-15 cell
line
C. Rhizome Dichloromethane extract Induces apoptosis [147]
purpurascens through mitochondrial-
dependent pathway in
colon cancer HT-29
cells
Essential oil Cytotoxicity against [148]
MCF7, Ca Ski, A549,
HT29, and HCT116
human carcinoma cell
lines
C. mutabilis Rhizome Petroleum ether extract and Induce apoptosis in [149]
labdane diterpenoid (Cm colorectal cancer
epoxide) HCT116 and leukemic
K562 cells
C. Rhizome Essential oil Cytotoxicity against [150]
kwangsiensis B16 and LNCaP cancer
cells
C. Rhizome Ethanol extract Antiproliferative [142]
phaeocaulis activity and induces
apoptosis in breast
cancer MCF7 cell lines
Genus Zingiber Boehmer
(continued)
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 439
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
Z. officinale Rhizome Ethanol extract Cytotoxicity against [151]
Human pancreatic
cancer cell lines
(Panc-1, AsPC-1,
BxPC-3, CAPAN-2,
CFPAC-1, MIAPaCa-2
and SW1990) as well
as induces autophagic
cell death in Panc 1
cells both in vitro and
in vivo (xenograft mice
model)
6-shogaol and 6-gingerol Induce apoptosis in [48]
against B164A5
murine melanoma cells
6-shogaol Induce cell cycle arrest [152]
and apoptosis against
in colon cancer cell
line HCT-116.
Antitumor activity
against colon cancer
cells (in vivo)
Induces endoplasmic [153]
reticulum stress and
mitochondrial
apoptosis induction in
cervical cancer HeLa
cells
Gingerol Induce apoptosis in [154]
SW-480 and HCT116
cancer cells
Induce apoptosis in [155]
A549 cells via extrinsic
pathway
Leaves Methanol extract Induce apoptosis in [156]
human colorectal
cancer HCT116 and
SW480 cells
(continued)
440 T. Soumya et al.
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
Z. zerumbet Rhizome Petroleum ether extract Cytotoxicity against [157]
fractions human breast cancer
MCF 7 cell lines
Zerumbone Induction of [158]
mitochondria-mediated
apoptosis in chronic
myelogenous leukemia
K562cells
Induces apoptosis in [159, 160]
CEM-ss, H1299,
HCT116, NB4,
P-388D1 and Raji
cancer cell lines.
Antitumor activity
against colorectal,
liver, lung and cervical
in vivo mice models
Z. Rhizome Chloroform extract Cytotoxicity against [161]
cassumunar Cis-3-(3’, human T-acute
4’-dimethoxyphenyl)-4-[(E)-3, lymphoblastic
4 dimethoxystyryl] cyclo-hex- leukemia (CEMss) and
1-ene and cervical (HeLa) cancer
8-(13,14-dimethoxyphenyl)-2- cell lines
methoxynaphto-1,4-quinone
Genus Elettaria Maton
E. Seeds Aqueous extract Cytotoxicity against [162]
cardamomum mouse lymphoma
YAC-1 cells.
Genus Hedychium J. Koenig
(continued)
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 441
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
H. Rhizome Ethanol extract Induce G1 phase cell [163]
coronarium arrest, apoptosis and
inhibition of migratory
potential of cervical
cancer HeLa cells
Labdane diterpenes Chemo preventive and [164]
antiproliferative
activity against HepG2
cancer cells
Cytotoxicity against
A-549, SK-N-SH,
MCF-7 and HeLa
cancer cell lines
Labdane diterpenoids Antiangiogenic activity [165]
(Hedycoronals A and B) and and cytotoxic activity
Diarylheptanoids against B16, HT-29,
HepG2 and HeLa
cancer cell lines
Coronarin D Induces G2/M arrest, [166]
apoptosis and
autophagy in human
nasopharyngeal
carcinoma NPC-BM
and NPC-039 cells
Induce apoptotic cell [167]
death through the
upregulation of JNK/
MAPK pathways in
human hepatocellular
carcinoma (HCC)
Huh7 and Sk-hep-1
cells
(continued)
442 T. Soumya et al.
Table 1 (continued)
Parts Bioactive extract / fractions /
Species name used compounds Anticancer activity References
H. spicatum Rhizome Labdane-diterpenoids Cytotoxic activity [168]
against Colo-205,
A-431, MCF-7, A549,
HL-60, THP-1, A-375
cancer cell lines and
Chinese hamster ovary
cells (CHO)
Sesquiterpenes Cytotoxicity against [169]
A549, B-16, HeLa,
HT-29, NCIH460,
PC-3, IEC-6 and L6
cancer cell lines
Essential oil Cytotoxicity against [170]
A549, DLD-1, SW
620, FaDu, HeLa
MCF-7 and
MDA-MB-231 cancer
cell lines
Genus Boesenbergia Kuntze
B. rotunda Rhizome Methanolic extract Cytotoxicity against [171]
MCF-7,
MDA-MB-231,
CaOV3, HT-29 and
HeLa cancer cell lines
Panduratin A Cytotoxicity and [172]
apoptosis induction
against HT-29 and
MCF-7 cancer cell
lines
Boesenbergin A Cytotoxicity against [173]
A549, PC3, HepG2
and HT-29 cancer cell
lines
Hexane and methanol extracts, Antiproliferative [174]
Cardamonin activity, induce cell
cycle arrest and
apoptosis against
nasopharyngeal
carcinoma, HK1 cells
Amomum is the second largest genus after Alpinia within Zingiberaceae with about
150 -180 species, widely distributed in Southeast Asia. In India, the genus is repre-
sented by 22 species, mostly restricted to North-Eastern and southern India. A chal-
cone, namely, cardamonin (2′,4′-dihydroxy-6′-methoxychalcone), first isolated
from A. subulatum (black cardamom) fruit, has been reported to affect cell growth
by modulation of a variety of cell signaling pathways, including mammalian target
of rapamycin (mTOR), NF-κB, cell surface receptors, and Wnt/β-catenin pathways
[180, 181]. Cardamonin also potentiates TNF-related apoptosis-inducing ligand
(TRAIL) for induction of apoptosis through ROS-CHOP (CCAAT/Enhancer-
Binding Protein Homologous Protein)-mediated upregulation of death receptors
(DRs), which makes TRAIL more effective as an anticancer therapy [91]. Hexane
and ethyl acetate extracts of A. subulatum seeds exhibited cytotoxicity against
MCF7 and HeLa cancer cell lines besides being immunosuppressive effect against
peripheral blood mononuclear cells [92]. Activity-guided fractionation of hexane-
and chloroform-soluble extracts of A. aculeatum leaves led to the isolation of acu-
leatin A and B, found to be cytotoxic against MCF7 breast cancer cell line (in vitro)
444 T. Soumya et al.
and in vivo hollow fiber assay [93]. A. villosum and A. kravanh have cytotoxic
effects against human hepatocellular carcinoma cell lines SMMC-7721 and Hep
G2, respectively [46, 96, 182]. Essential oil and various pure compounds such as
tsaokoarylone, isotsaokoin, hannokinol, 2,3-dihydro-2-(4′-hydroxy-phenylethyl)-6-
[(3″,4″-dihydroxy-5″-methoxy)phenyl]-4-pyrone, and 4-dihydro-2-(4′-hydroxy-
phenylmethyl)-6-[(3″,4″-dihydroxy-5″-methoxyphenyl)
methylene]-pyran-3,5-dione isolated from A. tsao-ko fruit (part of traditional
Chinese medicine) have been reported with cytotoxicity against various human can-
cer cell lines [94–96]. Ethanol extract from this plant was found to inhibit ovarian
cancer and decrease angiogenesis in vivo, through endoplasmic reticulum (ER)
stress-mediated interruption in p-STAT3/NF-κB/IL-6 and VEGF loop of angiogen-
esis regulation [97].
4.3 Genus Kaempferia L.
The genus includes about 70 species, two third of which are found in Asia and
remaining one third in Africa [33]. The flavanone, pinostrobin, isolated from
Kaempferia rotunda rhizome chloroform extract, has been shown to possess anti-
cancer activity against human breast cancer in vitro (T47D cell line) and in vivo
(Xenograft model). Repair of breast tissue and suppression of c-Myc expression
were evident on mice with T47D breast cancer xenograft [105]. Lectin isolated from
K. rotunda was found to inhibit proliferation of Ehrlich ascites carcinoma cells and
induce mitochondrial apoptosis in colorectal cancer cells, SW48 and SW480 [106,
107]. Alcoholic extracts of K. galanga rhizome were reported to possess antineo-
plastic activities in both in vivo and in vitro model systems [108, 109]. Interestingly,
water-soluble polysaccharides purified from K. galanga rhizome reportedly protect
thymus and spleen of solid tumor bearing mice and also capable of enhancing
immunoregulatory ability of CD4+ T cells, the cytotoxic effects of CD8+ T cells and
NK cells, thereby inhibiting tumor [110]. Additionally, major constituent of volatile
oil of K. galangal, Ethyl-p-methoxycinnamate, has been reported with anticancer
potential against oral cancer HSC-3 and Ca922 cell lines [111]. Ethanol extract of
K. parviflora rhizome, commonly known as Thai black ginger used in traditional
medicine, showed dose-dependent inhibition of cell proliferation and induction of
apoptosis in leukemic HL60 and U937 cell lines [112]. The ethanol extracts of
K. Parviflora rhizome supercritical CO2 fluid extracts (SFEs) of K. parviflora have
been contained in higher concentration of polymethoxyflavones (PMFs), which
showed potent antiproliferative activity against both human cervical (HeLa) and
gastric adenocarcinoma (AGS) cell lines [115]. It is also reported to possess anti-
cancer properties as evidenced by suppression of growth and survival signaling
pathways, inhibition of metalloproteinase 2 activity, inhibition of cell migration,
and invasion and induction of apoptosis in cancer cell lines such as HeLa (cervical)
and SKOV3 (ovarian) cells [113, 114]. Extracts and flavone derivatives from the
rhizome of K. parviflora have been shown to suppress multidrug
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 445
4.4 Genus Curcuma L.
The genus Curcuma L., with around 120 species, is distributed mainly in tropical
and subtropical Asia. Curcuma longa, commonly known as turmeric and a major
source of curcumin, has been consumed as a dietary spice and a cure for human ail-
ments for thousands of years in Asian countries. The potential anticancer activity of
turmeric and curcumin was demonstrated by Kuttan et al. 1985 [185] in both in vitro
and in vivo models. Various crude extracts of C. longa were reported to have antip-
roliferative activity against different human cancer cell lines such as, U937 (myeloid
leukemia), Molt4 (acute lymphoblastic leukemia), A549 (lung carcinoma), T98G
(glioblastoma), HeLa (cervical cancer), MDA-MB-231 (breast cancer), and murine
melanoma cell line (B164A5) [119–122]. The immunomodulatory activities of the
polar fractions of C. longa hot water extracts were investigated using human periph-
eral blood mononuclear cells (PBMC). High polarity fraction containing polysac-
charides exhibited stimulatory effects on PBMC proliferation, thereby attesting to
its potential use as an adjuvant supplement for cancer patients with suppressed
immunity due to exposure to chemotherapeutic drugs [186].
Other Curcuma sp. reported with antiproliferative potential include C. amada
(mango ginger), C. aromatica (wild turmeric), C. caesia (black turmeric), C. zedo-
aria (white turmeric), and C. xanthorrhiza (Java turmeric). Supercritical CO2 extract
of C. amada rhizome reported to have specific anticancer potential against human
glioblastoma (U-87MG) cell line induces apoptosis or drug resistance in a dose-
dependent manner [129]. Methanol extracts from C. amada leaves and rhizome also
reportedly possess antiproliferative activity against breast cancer cell lines MCF7
and MDA-MB-231 [130].
Aqueous extract of C. aromatica inhibited LS-174-T (colon carcinoma) cell pro-
liferation in a dose- and time-dependent manner, inducing extrinsic and intrinsic
apoptosis by activation of caspase-8, -9, and -3 and G2/M phase arrest.
Downregulation of cyclin B1 and CDK1 without the participation of p53 was also
observed in a study by Hu et al. [131]. Treatment with C. aromatica oil also inhib-
ited growth of implanted hepatoma in mice models which could be correlatable with
446 T. Soumya et al.
various cancer cell lines and induce apoptosis in colorectal cancer HCT116 and
leukemic K562 cells [149]. Essential oil isolated from various Curcuma species,
such as C. elata, C. kwangsiensis, C. yunnanensis, C. nankunshanensis, C. sichua-
nensis, C. rubescens, C. purpurascens, and C. mutabilis, also exhibited cytotoxicity
against various cell lines [148, 150, 190, 191].
Curcumin : Anticancer activity of curcumin need a special mention, as it’s a most
studied compound from Curcuma species of Zingiberaceae family. Curcuminoids
represent a major component of the phytoconstituents found in various Curcuma
species [192]. Of the various curcuminoids known, curcumin (1,7-bis(4-hydroxy-3-
methoxyphenyl)-1,6-heptadiene-3,5-dione), also called iferuloylmethane, deserves
a special mention. It is one of the most studied curcuminoids displaying a wide
spectrum of biological actions, including cholesterol-lowering, chemopreventive,
antidiabetic, anti-inflammatory, antimicrobial, and antioxidant activities [193–195].
Other commonly found curcuminoids are derivatives of curcumin which are known
as demethoxycurcumin and bisdemethoxycurcumin [196].
Curcumin has been studied in multiple human carcinomas including melanoma,
head and neck, breast, colon, pancreatic, prostate, and ovarian cancers [123–128].
Curcumin’s potent antioxidant and free-radical quenching properties play an impor-
tant role in the inhibitory effects of the compound on the initial stages of carcino-
genesis as demonstrated by animal models of various tumor types [197]. NF-κB and
AP-1 are two transcription factors intimately involved in the cellular pathways lead-
ing to tumorigenesis. NF-κB and AP-1 expression is induced by various stressful
stimuli (tumor promoters including oxidative stress, UV irradiation and infectious
antigens, pro-inflammatory cytokines such as TNF-α and IL-1), resulting in expres-
sion of genes involved in inflammation and cellular proliferation [198]. Curcumin
has an inhibitory effect on both NF-κB and AP-1 activation. Its effect on NF-κB, is
mediated through inhibition of IκK and results in inactive NF-κB remaining bound
to IκBα in the cytoplasm leading to suppression of a variety of gene products
involved in carcinogenesis and tumor growth including cyclin D1, VEGF (Vascular
endothelial growth factor), COX-2 (cycloxygenase-2), c-myc, Bcl-2, ICAM-1, and
MMP-9 (Matrix metalloproteinase-9) [199]. Curcumin also has a stimulatory effect
on the extrinsic apoptotic pathway, which is triggered by the binding of ‘death acti-
vators’ such as TNF-α and Fas-ligand to their corresponding cell surface receptors.
In addition to proapoptotic effect, curcumin also induces autophagic cell death in
chronic myelogenous leukemia, esophageal cancer, and malignant glioma cells,
mediated through inhibition of the Akt/mTOR/p70S6 kinase pathway and the
ERK1/2 pathway [200, 201]. Curcumin has demonstrated antiangiogenic effect in
vivo xenograft models, by regulating a variety of proangiogenic growth factors,
enzymes, and transcription factors like bFGF (basic fibroblast growth factor),
VEGF, angiopoetin-1 and 2, COX-2 MMP-9 [126, 202]. Its derivative, demethoxyc-
urcumin (DMC), has been reported to affect a number of cellular adhesion mole-
cules involved in the processes of metastasis [203].
Curcumin is known to target mTOR, which is recognized as a key therapeutic
target for the prevention and / or treatment of cancer [204]. Curcumin has been
shown to have numerous cytotoxic effects on cancer stem cells (CSCs) by
448 T. Soumya et al.
suppressing the release of cytokines, particularly interleukin (IL)-6, IL-8, and IL-1,
which stimulate CSCs [205]. It is an inhibitor of enzymes involved in epigenetic
changes of chromatin such as DNA methyltransferase, histone acetyl transferase,
and histone deacetylase (HDAC) leading to selective activation or inactivation of
genes (oncogenes/tumor suppressors) implicated in cancer death and progression.
Curcumin also modulates miRNAs (miR-15a, miR-16, miR-21, miR-22, miR-26,
miR-101, miR-146, miR-200, miR-203, and let-7) and their multiple target genes.
Altogether, curcumin is able to restore the epigenetic regulation balance and appears
as an attractive preventive and/or therapeutic approach against human cancer
[206, 207].
Although curcumin has long been used extensively to treat several inflammatory
diseases including cancer, poor aqueous solubility and reduced bioavailability limit
its efficacy as a promising therapeutic agent in cancer therapy. Various research
groups have focused on increasing the bioavailability of curcumin by combining
other phytochemicals as adjuvants. For instance, curcumin has often been used in
combination with other phytochemicals such as resveratrol, quercetin, sulfora-
phane, retinoic acid, and folates in cancer treatment [208–210]. The chemosensitiz-
ing effect of curcumin has been reported in cancers of the breast, colon, pancreas,
gut, liver, lung, prostate, brain, lymphoma, and leukemia [211, 212]. Various types
of curcumin nanoparticles appropriate for cancer treatment have been developed,
such as polymer nanoparticles, liposomes, micelles, solid lipid nanoparticles
(SLNs), and polymer conjugates, with improved bioavailability, devoid of degrada-
tion and further metabolism and with enhanced targeting capacities [207, 213, 214].
5 Conclusion
References
1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A (2018) Global cancer statis-
tics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185
countries. CA Cancer J Clin. 68:394–424. http://www.ncbi.nlm.nih.gov/pubmed/30207593
2. Ruddon RW (2007) Cancer biology, 4th edn. Oxford University Press
3. Anand P, Kunnumakara AB, Sundaram C, Harikumar KB, Tharakan ST, Lai OS, Sung B,
Aggarwal BB (2008) Cancer is a preventable disease that requires major lifestyle changes.
Pharm Res 25:2097–2116
4. Cohen L, Jefferies A (2017) Comprehensive lifestyle change: Harnessing synergy to improve
cancer outcomes. J Natl Cancer Inst. 2017:33–36
5. Weinberg RA (2014) The Biology of Cancer, 2nd edn. Garland Science, Taylor and
Francis group
6. Ke X, Shen L (2017) Molecular targeted therapy of cancer: The progress and future prospect.
Front Lab Med 1:69–75. https://doi.org/10.1016/j.flm.2017.06.001
7. Kooti W, Servatyari K, Behzadifar M, Asadi-Samani M, Sadeghi F, Nouri B, Zare MH (2017)
Effective medicinal plant in cancer treatment. J Evidence-Based Complement Altern Med
22(4):982–995
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 451
8. Thapliyal A, Khar RK, Amrish ChandraChandra A (2018) Overview of cancer and medicinal
herbs used for cancer therapy. Asian J Pharm 12:1–8. https://www.asiapharmaceutics.info/
index.php/ajp/article/view/2033
9. Guerra B, Issinger O-G (2019) Natural compounds and derivatives as Ser/Thr protein kinase
modulators and inhibitors. Pharmaceuticals 12:4. https://doi.org/10.3390/ph12010004
10. Tariq A, Sadia S, Pan K, Ullah I, Mussarat S, Sun F, Abiodun OO, Batbaatar A, Li Z, Song
D, Xiong Q, Ullah R, Khan S, Basnet BB, Kumar B, Islam R, Adnan M (2017) A systematic
review on ethnomedicines of anti-cancer plants. Phyther Res 31:202–264
11. Seca AML, Pinto DCGA (2018) Plant secondary metabolites as anticancer agents: Successes
in clinical trials and therapeutic application. Int J Mol Sci 19(1):263
12. Cragg GM, Pezzuto JM (2016) Natural products as a vital source for the discovery of cancer
chemotherapeutic and chemopreventive agents. Med Princ Pract 25:41–59
13. Iqbal J, Abbasi BA, Kanwal S, Khalil AT, Mahmood T, Shah SA, Ali B (2017) Plant-derived
anticancer agents: a green anticancer approach. Asian Pac J Trop Biomed 7:1129–1150.
https://doi.org/10.1016/j.apjtb.2017.10.016
14. Kress WJ, Prince LM, Williams KJ (2002) The phylogeny and a new classification of the
gingers (Zingiberaceae): evidence from molecular data. Am J Bot 89(11):1682–1696
15. Sabu M (2006) Zingiberaceae and Costaceae of South India. Indian Association for
Angiosperm Taxonomy
16. Pancharoen O, Prawat U, Tuntiwachwuttikul P (2000) Phytochemistry of the Zingiberaceae.
Stud Nat Prod Chem 23:797–865
17. Fadilah F, Yanuar A, Arsianti A, Andrajati R (2017) Phenylpropanoids, eugenol scaffold, and
its derivatives as anticancer. Asian J Pharm Clin Res 10:41–46
18. Ansari IA, Akhtar MS (2019) Chapter 3 - Current insights on the role of terpenoids as anti-
cancer agents: a perspective on cancer prevention and treatment. In: Swamy MK, Akhtar
MS (eds) Nature Bio-active Compound. Springer Nature Singapore, pp 53–80. https://doi.
org/10.1007/978-981-13-7205-6_3
19. Kopustinskiene DM, Jakstas V, Savickas A, Bernatoniene J (2020) Flavonoids as anticancer
agents. Nutrients 12:457. https://doi.org/10.3390/nu12020457
20. Basak S, Sarma GC, Rangan L (2010) Ethnomedical uses of Zingiberaceous plants of
Northeast India. J Ethnopharmacol 132:286–296
21. Lakshmi S, Padmaja G, Remani P (2011) Antitumour effects of Isocurcumenol isolated from
Curcuma zedoaria rhizomes on human and murine cancer cells. Int J Med Chem. https://doi.
org/10.1155/2011/253962
22. Prasad S, Tyagi AK (2015) Ginger and its constituents: role in prevention and treatment of
gastrointestinal cancer. Gastroenterol Res Pract. https://doi.org/10.1155/2015/142979
23. Chen D, Li H, Li W, Feng S, Deng D (2018) Kaempferia parviflora and its Methoxyflavones:
chemistry and biological activities. Evidence-Based Complement Altern Med. https://doi.
org/10.1155/2018/4057456
24. Alkandahri MY, Shafirany MZ, Rusdin A, Agustina LS, Pangaribuan F, Fitrianti F, Farhamzah
KAH, Sugiharta S, Mardiana LA (2021) Amomum compactum: a review of pharmacological
studies. Plant Cell Biotechnol Mol Biol 22:61–69
25. Kirana C, Record IR, McIntosh GH, Jones GP (2003) Screening for antitumor activity of 11
species of Indonesian Zingiberaceae using human MCF-7 and HT-29 cancer cells. Pharm
Biol 41:271–276
26. Afzal A, Oriqat G, Khan AM, Jose J, Afzal M (2013) Chemistry and biochemistry of terpe-
noids from Curcuma and related species. J Biol Active Prod Nat 3:1–55. http://www.tandfon-
line.com/doi/abs/10.1080/22311866.2013.782757
27. Ghosh S, Rangan L (2013) Alpinia: the gold mine of future therapeutics. 3 Biotech 3:173–185
28. Hartati R, Suganda AG, Fidrianny I (2014) Botanical, phytochemical and pharmacological
properties of Hedychium (Zingiberaceae) – a review. Procedia Chem 13:150–163. http://link-
inghub.elsevier.com/retrieve/pii/S1876619614002095
452 T. Soumya et al.
29. Wallace D (2016) Natural products as a source of anti-cancer lead compounds: Ginger and
breast cancer. J Pharmacol Clin Res 1(3):1–5
30. Joy PP, Thomas J, Mathew S, Skaria BP (1998) Zingiberaceous medicinal and aromatic
plants. Aromatic and Medicinal Plants Research Station, Odakkali
31. Sirirugsa P (1998) Thai Zingiberaceae: Species diversity and their uses. Pure Appl Chem
70:23–27
32. Jatoi SA, Kikuchi A, Watanabe KN (2007) Genetic diversity, cytology, and systematic and
phylogenetic studies in Zingiberaceae fleshy roots. Genes Genome Genom 1(1):56–62
33. Prabhu KKM, Asish G, Sabu M, Balachandran I (2013) Significance of gingers (Zingiberaceae)
in indian system of medicine - Ayurveda: an overview. Anc Sci Life 32:253
34. Zahara M, Hasanah M, Zalianda R (2018) Identification of Zingiberaceae as medicinal plants
in Gunung cut village, Aceh Barat Daya, Indonesia. J Trop Hortic. 1:24–28
35. Chhabra SC, Mahunnah RLA, Mshiu EN (1993) Plants used in traditional medicine in Eastern
Tanzania. VI. Angiosperms (Sapotaceae to Zingiberaceae). J Ethnopharmacol 39:83–103
36. Peng L, Zou HQ, Bauer R, Liu Y, Tao O, Yan SR, Han Y, Li JH, Ren ZY, Yan YH (2014)
Identification of Chinese herbal medicines from Zingiberaceae family using feature extraction
and cascade classifier based on response signals from E-Nose. Evidence-based Complement
Altern Med. https://doi.org/10.1155/2014/963035
37. Ujang Z, Subramaniam T, Nordin NI (2015) Ginger species and their traditional uses in mod-
ern applications. J Ind Technol 23:59–70
38. Kasarkar AR, Kulkarni DK (2016) Traditional knowledge of medicines belonging to family
Zingiberaceae from South Western Maharashtra, India. Int J Bot Stud 1(4):20–23
39. Jantan IB, Yassin MSM, Chin CB, Chen LL, Sim NL (2003) Antifungal activity of the essen-
tial oils of nine Zingiberaceae species. Pharm Biol 41(5):392–397
40. Cheenpracha S, Karalai C, Ponglimanont C, Subhadhirasakul S, Tewtrakul S (2006) Anti-
HIV-1 protease activity of compounds from Boesenbergia pandurata. Bioorganic Med Chem
14:1710–1714
41. Tewtrakul S, Subhadhirasakul S (2007) Anti-allergic activity of some selected plants in the
Zingiberaceae family. J Ethnopharmacol 109:535–538
42. Chen IN, Chang CC, Ng CC, Wang CY, Shyu YT, Chang TL (2008) Antioxidant and antimi-
crobial activity of Zingiberaceae plants in Taiwan. Plant Foods Hum Nutr 63:15–20
43. Hanish Singh JC, Alagarsamy V, Diwan PV, Sathesh Kumar S, Nisha JC, Narsimha RY
(2011) Neuroprotective effect of Alpinia galanga (L.) fractions on Aβ(25–35) induced amne-
sia in mice. J Ethnopharmacol 138:85–91
44. Kalaivani K, Senthil-Nathan S, Murugesan AG (2012) Biological activity of selected
Lamiaceae and Zingiberaceae plant essential oils against the dengue vector Aedes aegypti
L. (Diptera: Culicidae). Parasitol Res 110:1261–1268
45. Salama SM, Abdulla MA, AlRashdi AS, Ismail S, Alkiyumi SS, Golbabapour S (2013)
Hepatoprotective effect of ethanolic extract of Curcuma longa on thioacetamide induced
liver cirrhosis in rats. BMC Compl Alter Med 13:56. http://www.biomedcentral.
com/1472-6882/13/56
46. Lu CL, Zhao HY, Jiang JG (2013) Evaluation of multi-activities of 14 edible species from
Zingiberaceae. Int J Food Sci Nutr 64(1):28–35
47. Al-Nahain A, Jahan R, Rahmatullah M (2014) Zingiber officinale: A potential plant
against rheumatoid arthritis. Evidence-Based Complement Altern Med. https://doi.
org/10.1155/2014/159089
48. Danciu C, Vlaia L, Fetea F, Hancianu M, Coricovac DE, Ciurlea SA, Şoica CM, Marincu I,
Vlaia V, Dehelean CA, Trandafirescu C (2015) Evaluation of phenolic profile, antioxidant
and anticancer potential of two main representants of Zingiberaceae family against B164A5
murine melanoma cells. Biol Res 48:1–9. http://www.biolres.com/content/48/1/1
49. Lakhan SE, Ford CT, Tepper D (2015) Zingiberaceae extracts for pain: a systematic review
and meta-analysis. Nutr J 14:50. https://doi.org/10.1186/s12937-015-0038-8
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 453
50. Nithya R, Jayshree N (2017) A review on herbs of the Zingiberaceae family with beneficial
effects on cardiovasular diseases. World J Pharm Pharm Sci 6:635–643
51. Aghasi M, Ghazi-Zahedi S, Koohdani F, Siassi F, Nasli-Esfahani E, Keshavarz A, Qorbani
M, Khoshamal H, Salari-Moghaddam A, Sotoudeh G (2018) The effects of green Cardamom
supplementation on blood glucose, lipids profile, oxidative stress, Sirtuin-1 and Irisin in type
2 diabetic patients: a study protocol for a randomized placebo-controlled clinical trial. BMC
Complement Altern Med 18:1–6. https://doi.org/10.1186/s12906-017-2068-6
52. Raju R, Singh A, Gunawardena D, Reddell P, Münch G (2019) Diarylheptanoids with anti-
inflammatory activity from the rhizomes of Pleuranthodium racemigerum (Zingiberaceae).
Phytochem Lett 30:10–13
53. Ganapathy G, Preethi R, Moses JA, Anandharamakrishnan C (2019) Diarylheptanoids as
nutraceutical: a review. Biocatal Agric Biotechnol 19:101109
54. Gurib-Fakim A, Maudarbaccus N, Leach D, Doimo L, Wohlmuth H (2002) Essential oil com-
position of Zingiberaceae species from Mauritius. J Essent Oil Res 14:271–273
55. Abe M, Nakamura Y, Yamada Y, Osawa T, Morimitsu Y, Uda Y (2003) Labdane-type diter-
pene dialdehyde, pungent principle of Myoga, Zingiber mioga Roscoe. Biosci Biotechnol
Biochem 66(12):2698–2700
56. Chimnoi N, Sarasuk C, Khunnawutmanotham N, Intachote P, Seangsai S, Saimanee B,
Pisutjaroenpong S, Mahidol C, Techasakul S (2009) Phytochemical reinvestigation of
labdane-type diterpenes and their cytotoxicity from the rhizomes of Hedychium coronarium.
Phytochem Lett 2:184–187
57. Manse Y, Ninomiya K, Nishi R, Kamei I, Katsuyama Y, Imagawa T, Chaipech S, Muraoka
O, Morikawa T (2016) Melanogenesis inhibitory activity of a 7-O-9’-linked neolignan from
Alpinia galanga fruit. Bioorganic Med Chem 24:6215–6224. https://doi.org/10.1016/j.
bmc.2016.10.001
58. Win NN, Ito T, Ngwe H, Win YY, Prema OY, Tanaka M, Asakawa Y, Abe I, Morita H (2017)
Labdane diterpenoids from Curcuma amada rhizomes collected in Myanmar and their antip-
roliferative activities. Fitoterapia 122:34–39
59. Ji KL, Fan YY, Ge ZP, Sheng L, Xu YK, Gan LS, Li JY, Yue JM (2019) Maximumins A-D,
rearranged Labdane-type diterpenoids with four different carbon skeletons from Amomum
maximum. J Org Chem 84:282–288
60. Sematong T, Pongprayoon U, Tuchinda P, Claeson P, Reutrakul V, Nahar N (1996) Topical
antiinflammatory activity of two pimarane diterpenes from Kaempferia pulchra. Phyther Res
10:534–535
61. Alberti Á, Riethmüller E, Béni S (2018) Characterization of diarylheptanoids: an emerging
class of bioactive natural products. J Pharm Biomed Anal. 147:13–34
62. Kim NJ, Byun SG, Cho JE, Chung K, Ahn YJ (2008) Larvicidal activity of Kaempferia
galanga rhizome phenylpropanoids towards three mosquito species. Pest Manag Sci
64:857–862
63. Kuddus R, Rumi F, Kaisar A, Hasan CM (2010) Sesquiterpene and phenylpropanoids from
Curcuma longa. Bangladesh Pharm J 13(2):31–34
64. Hong SS, Oh JS (2012) Phenylpropanoid ester from Zingiber officinale and their inhibitory
effects on the production of nitric oxide. Arch Pharm Res 35:315–320
65. Samarghandian S, Hadjzadeh MAR, Afshari JT, Hosseini M (2014) Antiproliferative activ-
ity and induction of apoptotic by ethanolic extract of Alpinia galanga rhizhome in human
breast carcinoma cell line. BMC Complement Altern Med 14:192. http://www.biomedcen-
tral.com/1472-6882/14/192
66. Chouni A, Paul S (2018) A review on phytochemical and pharmacological potential of
Alpinia galanga. Pharmacogn J 10(1):9–15
67. Rao CH, Namosiva T, Suryaprakasam S (1976) Cardamonin and Alpinetin from the seeds of
Amomum subulatum. Planta Med 29:391–392
68. Jang DS, Han A-R, Park G, Jhon G-J, Seo E-K (2004) Flavonoids and aromatic compounds
from the rhizomes of Zingiber zerumbet. Arch Pharm Res 27(4):386–389
454 T. Soumya et al.
69. Ching AYL, Wah TS, Sukari MA, Lian GEC, Rahmani M, Khalid K (2007) Characterization
of flavonoid derivatives from Boesenbergia rotunda (L.). Malay J Anal Sci 11:154–159
70. Sutthanut K, Sripanidkulchai B, Yenjai C, Jay M (2007) Simultaneous identification and
quantitation of 11 flavonoid constituents in Kaempferia parviflora by gas chromatography. J
Chromatogr A 1143:227–233
71. Liu D, Qu W, Liang JY (2013) Flavonoids and other constituents from Alpinia sichuanensis
Z.Y. Zhu. Biochem Syst Ecol 46:127–129
72. Sabulal B, Dan M, John JA, Kurup R, Purushothaman CS, George V (2007) Phenylbutanoid-
rich rhizome oil of Zingiber neesanum from Western Ghats, Southern India. Flavour Fragr J
22:521–524
73. Taechowisan T, Suttichokthanakorn S, Phutdhawong WS (2018) Antibacterial and cyto-
toxicity activities of phenylbutanoids from Zingiber cassumunar Roxb. J Appl Pharm Sci
8:121–127
74. He ZH, Ge W, Yue GGL, Lau CBS, He MF, But PPH (2010) Anti-angiogenic effects of the
fruit of Alpinia oxyphylla. J Ethnopharmacol 132:443–449
75. Nam JW, Kim SJ, Han RM, Lee SK, Seo EK (2005) Cytotoxic phenylpropanoids from the
rhizome of Alpinia galanga. J Appl Pharm 13:263–266
76. Awang K, Nurul Azmi M, Lian Aun LI, Nazif Aziz A, Ibrahim H, Hasima NN (2010) The
apoptotic effect of 1’S-1’-Acetoxychavicol acetate from Alpinia conchigera on human can-
cer cells. Molecules 15:8048–8059
77. Lu Y, Wang Z, Wei D, Xiang H (2007) Mechanism and inhibitory effect of galangin and its
flavonoid mixture from Alpinia officinarum on mushroom tyrosinase and B16 murine mela-
noma cells. J Enz Inhibit Med Chem 22(4):433–438
78. An N, Zou Z, Tian Z, Luo X, Yang S, Xu L (2008) Diarylheptanoids from the rhizomes of
Alpinia officinarum and their anticancer activity. Fitoterapia 79:27–31
79. Tabata K, Yamazaki U, Okada M, Fukumura K, Shimada A, Sun Y, Yasukawa K, Suzuki T
(2009) Diarylheptanoids derived from Alpinia officinarum induce apoptosis, S-phase arrest
and differentiation in human neuroblastoma cells. Anticancer Res 29:4981–4988
80. Malek ANS, Phang CW, Ibrahim H, Wahab NA, Sim KS (2011) Phytochemical and cytotoxic
investigations of Alpinia mutica rhizomes. Molecules 16:583–589. https://doi.org/10.3390/
molecules16010583
81. Raj CA, Ragavendran P, Sophia D, Rathi MA, Gopalakrishnan VK (2012) Evaluation of in-
vitro antioxidant and anticancer activity of Alpinia purpurata. Chin J Nat Med 10(4):263–268
82. Oirere EK, Anusooriya P, Malarvizhi D, Raj CA, Gopalakrishnan VK (2016) Antioxidant,
cytotoxic and apoptotic activities of crude extract of Alpinia purpurata on cervical cancer cell
line. Int J Pharm Sci Rev Res 36(2):28–34
83. Phang C, Nurestri S, Malek A, Ibrahim H (2013) Antioxidant potential, cytotoxic activity
and total phenolic content of Alpinia pahangensis rhizomes. BMC Complement Altern Med
13:243. http://www.biomedcentral.com/1472-6882/13/243
84. Sim KS, Ibrahim H, Malek ANS, Syamsir DR, Awang K (2014) Cytotoxic activity of Alpinia
murdochii Ridl: a mountain ginger species from Peninsular Malaysia. Pharmaco Mag
10:70–72
85. Hua SZ, Luo JG, Wang XB, Wang JS, Kong LY (2009) Two novel monoterpene-chalcone
conjugates isolated from the seeds of Alpinia katsumadai. Bioorganic Med Chem Lett
19:2728–2730. https://doi.org/10.1016/j.bmcl.2009.03.117
86. Zahra MH, Salem TAR, El-Aarag B, Yosri N, EL-Ghlban S, Zaki K, Marei AH, EL-Wahed
AA, Saeed A, Khatib A, AlAjmi MF, Shathili AM, Xiao J, Khalifa SAM, El-Seedi HR (2019)
Alpinia zerumbet (Pers.): Food and medicinal plant with potential in vitro and in vivo anti-
cancer activities. Molecules 24:2495. https://doi.org/10.3390/molecules24132495
87. Kuo C-Y, Teng-Song Weng T-S, Senthil Kumar KJ, Tseng Y-H, Tung T-W, Wang S-Y,
Wang H-C (2019) Ethanol Extracts of Dietary Herb, Alpinia nantoensis, exhibit anti-
cancer potential in human breast cancer cells. Integr Cancer Ther 18:1–12. https://doi.
org/10.1177/153473541986692
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 455
88. Reddy AS, Abd Malek SN, Ibrahim H, Sim KS (2013) Cytotoxic effect of Alpinia scabra
(Blume) Náves extracts on human breast and ovarian cancer cells. BMC Complement Altern
Med 13:314. https://doi.org/10.1186/1472-6882-13-314
89. Ali MS, Banskota AH, Tezuka Y, Saiki I, Kadota S (2001) Antiproliferative activity of diaryl-
heptanoids from the seeds of Alpinia blepharocalyx. Biol Pharm Bull 24(5):525–528
90. Yang HL, Chen SC, Chen CS, Wang SY, Hseu YC (2008) Alpinia pricei rhizome extracts
induce apoptosis of human carcinoma KB cells via a mitochondria-dependent apoptotic path-
way. Food Chem Toxicol 46:3318–3324
91. Yadav VR, Prasad S, Aggarwal BB (2012) Cardamonin sensitizes tumour cells to TRAIL
through ROS- and CHOP- mediated up-regulation of death receptors and down- regulation of
survival. Brit J Pharma 165:741–753
92. Sharma V, Lohia N, Handa V, Baranwal M (2017) Amomum subulatum seed extract exhibit
antioxidant, cytotoxic and immune-suppressive effect. Indian J Biochem Biophys 54:135–139
93. Chin YW, Salim AA, Su BN, Mi Q, Chai HB, Riswan S, Kardono LBS, Ruskandi A,
Farnsworth NR, Swanson SM, Kinghorn AD (2008) Potential anticancer activity of naturally
occurring and semisynthetic derivatives of aculeatins A and B from Amomum aculeatum. J
Nat Prod 71:390–395
94. Moon SS, Cho SC, Lee JY (2005) Tsaokoarylone, a cytotoxic diarylheptanoid from Amomum
tsao-ko fruits. Bull Korean Chem Soc 26:447–450
95. Yang Y, Yue Y, Runwei Y, Guolin Z (2010) Cytotoxic, apoptotic and antioxidant activity of
the essential oil of Amomum tsao-ko. Bioresour Technol 101:4205–4211
96. Zhang T-T, Lu C-L, Jiang J-G (2015) Antioxidant and anti-tumour evaluation of compounds
identified from fruit of Amomum tsaoko Crevost et Lemaire. J Funct Foods. 18:423–431
97. Chen C, You F, Wu F, Luo Y, Zheng G, Xu H, Liu Y (2020) Antiangiogenesis Efficacy of
Ethanol Extract from Amomum tsaoko in Ovarian Cancer through Inducing ER Stress to
Suppress p-STAT3/NF-kB/IL-6 and VEGF Loop. Evidence-based Complement Altern
Med:2390125. https://doi.org/10.1155/2020/2390125
98. Zhang D, Li S, Xiong Q, Jiang C, Lai X (2013) Extraction, characterization and biological
activities of polysaccharides from Amomum villosum. Carbohydr Polym 95:114–122
99. Tangjitjaroenkun J, Tangchitcharoenkhul R, Yahayo W, Supabphol S, Sappapan R, Supabphol
R (2020) Chemical compositions of essential oils of Amomum verum and Cinnamomum par-
thenoxylon and their in vitro biological properties. J Herbmed Pharmacol 9(3):223–231
100. Choi JW, Kim KH, Lee IK, Choi SU, Lee KR (2009) Phytochemical constituents of Amomum
xanthioides. Nat Prod Sci 15(1):44–49
101. Kim KH, Choi JW, Choi SU, Lee KR (2010a) Terpene glycosides and cytotoxic constituents
from the seeds of Amomum xanthioides. Planta Med 76(5):461–464
102. Kim KH, Choi JW, Choi SU, Seo EK, Lee KR (2010b) Amoxantin A: a new bisnorlabdane
diterpenoid from Amomum xanthioides. Bull Kor Chem Soc 31(4):1035–1037
103. Kim KH, Choi JW, Choi SU, Lee K (2011) Cytotoxic sesquiterpenoid from the seeds of
Amomum xanthioides. Nat Prod Sci 17(1):10–13
104. Luo JG, Yin H, Fan BY, Kong LY (2014) Labdane diterpenoids from the roots of Amomum
maximum and their cytotoxic evaluation. Helv Chim Acta 97(8):1140–1145
105. Atun S, Arianingrum R (2015) Anticancer activity of bioactive compounds from Kaempferia
rotunda rhizome against human breast cancer. Int J Pharmacogn Phytochem Res 7:262–269
106. Kabir SR, Reza MA (2014) Antibacterial activity of Kaempferia rotunda rhizome lectin
and its induction of apoptosis in Ehrlich ascites carcinoma cells. Appl Biochem Biotechnol
172:2866–2876
107. Islam F, Gopalan V, Lam AKY, Kabir SR (2019) Kaempferia rotunda tuberous rhizome lectin
induces apoptosis and growth inhibition of colon cancer cells in vitro. Int J Biol Macromol
141:775–782. https://doi.org/10.1016/j.ijbiomac.2019.09.051
108. Amuamuta A, Plengsuriyakarn T, Na-Bangchang K (2017) Anticholangiocarcinoma activity
and toxicity of the Kaempferia galanga Linn. Rhizome ethanolic extract. BMC Complement
Altern Med 17:213. https://doi.org/10.1186/s12906-017-1713-4
456 T. Soumya et al.
109. Ali H, Yesmin R, Satter Mohammed A, Habib R, Yeasmin T (2018) Antioxidant and antineo-
plastic activities of methanolic extract of Kaempferia galanga Linn. Rhizome against Ehrlich
ascites carcinoma cells. J King Saud Univ Sci 30:386–392
110. Yang X, Ji H, Feng Y, Yu J, Liu A (2018) Structural characterization and antitumor activity of
polysaccharides from Kaempferia galanga L. Oxid Med Cell Longev:9579262. https://doi.
org/10.1155/2018/9579262
111. Ichwan SJA, Husin A, Suriyah WH, Lestari W, Omar MN, Kasmuri AR (2019) Anti-
neoplastic potential of ethyl-p-methoxycinnamate of Kaempferia galanga on oral cancer cell
lines. Mater Today Proc 16:2115–2121. https://doi.org/10.1016/j.matpr.2019.06.100
112. Banjerdpongchai R, Chanwikruy Y, Rattanapanone V, Sripanidkulchai B (2009) Induction
of apoptosis in the human leukemic U937 cell line by Kaempferia parviflora Wall.Ex.Baker
extract and effects of Paclitaxel and Camptothecin. Asian Pac J Cancer Prev 10:1137–1140
113. Potikanond S, Sookkhee S, Takuathung MN, Mungkornasawakul P, Wikan N, Smith DR,
Nimlamool W (2017) Kaempferia parviflora extract exhibits anti-cancer activity against
HeLa cervical cancer cells. Front Pharmacol 8:630. https://doi.org/10.3389/fphar.2017.00630
114. Paramee S, Sookkhee S, Sakonwasun C, Takuathung MN, Mungkornasawakul P,
Nimlamool W, Potikanond S (2018) Anti-cancer effects of Kaempferia parviflora on ovar-
ian cancer SKOV3 cells. BMC Complement Altern Med 18:178. https://doi.org/10.1186/
s12906-018-2241-6
115. Wongsrikaew N, Kim H, Vichitphan K, Cho SK, Han J (2012) Antiproliferative activity and
polymethoxyflavone composition analysis of Kaempferia parviflora extracts. J Korean Soc
Appl Biol Chem 55:813–817
116. Leardkamolkarn V, Tiamyuyen S, Sripanidkulchai BO (2009) Pharmacological activity of
Kaempferia parviflora extract against human bile duct cancer cell lines. Asian Pac J Cancer
Prev 10:695–698
117. Tang SW, Sukari MA, Neoh BK, Yeap YSY, Abdul AB, Kifli N, Cheng Lian Ee G (2014)
Phytochemicals from Kaempferia angustifolia Rosc. and their cytotoxic and antimicrobial
activities. Biomed Res Int. https://doi.org/10.1155/2014/417674
118. Chawengrum P, Boonsombat J, Kittakoop P, Mahidol C, Ruchirawat S, Thongnest S (2018)
Cytotoxic and antimicrobial labdane and clerodane diterpenoids from Kaempferia elegans
and Kaempferia pulchra. Phytochem Lett 24:140–144
119. Kaneshiro T, Suzui M, Takamatsu R, Murakami A, Fujino T, Yoshimi N (2005) Growth inhib-
itory activities of crude extracts obtained from herbal plants in the Ryukyu Islands on several
human colon carcinoma cell lines. Asian Pac J Cancer Prev 6:353–358
120. Ahmad R, Srivastava AN, Khan MA (2016) Evaluation of in-vitro anticancer activity of rhi-
zome of Curcuma longa against human breast cancer and Vero cell lines. Int J Bot Stud 1:1–6
121. Hadem KLH, Sen A (2017) Curcuma species: a source of anticancer drugs. J Tumor Med
Prev 1(5):1–7
122. Kukula-Koch W, Grabarska A, Jarogniew Ł, Czernicka L, Nowosadzka E, Gumbarewicz E,
Jarzab A, Audo G, Upadhyay S, Głowniak K, Stepulak A (2018) Superior anticancer activity
is demonstrated by total extract of Curcuma longa L. as opposed to individual curcuminoids
separated by centrifugal partition chromatography. Phytother Res 32:933–942
123. Liu D, Chen Z (2013) Breast cancer the effect of curcumin on breast cancer cells. J Breast
Can 16(2):133–137
124. Mukhopadhyay A, Bueso-ramos C, Chatterjee D, Pantazis P, Aggarwal BB (2001) Curcumin
downregulates cell survival mechanisms in human prostate cancer cell lines. Oncogene
20:7597–7609
125. Hanif R, Qiao L, Shiff SJ, Rigas B (1997) Curcumin, a natural plant phenolic food additive,
inhibits cell proliferation and induces cell. J Lab Clin Med 130(6):576–584
126. Lin YG, Kunnumakkara AB, Nair A, Merritt WM, Han L, Armaiz-pena GN, Kamat AA,
Spannuth W, Gershenson DM, Lutgendorf SK, Aggarwal BB, Sood AK (2007) Curcumin
inhibits tumor growth and angiogenesis in ovarian carcinoma by targeting the Nuclear
Factor-κB pathway. Clin Cancer Res 13(11):3423–3431
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 457
127. Siwak DR, Shishodia S, Aggarwal BB, Kurzrock R (2005) Curcumin-induced antiprolifera-
tive and proapoptotic effects in melanoma cells are associated with suppression of IκB kinase
and Nuclear Factor-κB activity and are independent of the B-Raf / mitogen- a ctivated / extra-
cellular signal-regulated protein kinase pathway and the Akt pathway. Cancer 104(5):879–890
128. Wang D, Veena MS, Stevenson K, Tang C, Ho B, Suh JD, Duarte VM, Faull KF, Mehta
K, Srivatsan ES, Wang MB (2008) Liposome-encapsulated Curcumin suppresses growth of
head and neck squamous cell carcinoma in vitro and in xenografts through the inhibition
of Nuclear Factor-κB by an AKT-independent pathway. Clin Cancer Res 14(19):6228–6237
129. Ramachandran C, Lollett IV, Escalon E, Quirin K, Melnick SJ (2015) Anticancer potential
and mechanism of action of Mango ginger (Curcuma amada Roxb.) supercritical CO2 extract
in human glioblastoma cells. J Evid-Based Compl Altern Med 20(2):109–119
130. Sivaprabha J, Dharani B, Padma PR, Sumathi S (2016) Apoptosis-induced in-vitro anticancer
activity of methanolic extract of leaves and rhizomes of Curcuma amada Roxb. against breast
cancer cells. Int J Green Pharm 10(2):98–103
131. Hu B, Shen K-P, An H-M, Wu Y, Du Q (2011) Aqueous extract of Curcuma aromatica
induces apoptosis and G2/M arrest in human colon carcinoma LS-174-T cells independent of
p53. Can Biother Radiopharm 26:97–104
132. Wu WY, Xu Q, Shi LC, Zhang WB (2000) Inhibitory effects of Curcuma aromatica oil on
proliferation of hepatoma in mice. World J Gastroentero 6(2):216–219
133. Li Y, Wo JM, Ms QL, Li X, Martin RCG (2009) Chemoprotective effects of Curcuma aro-
matica on esophageal carcinogenesis. Ann Surg Oncol 16:515–523
134. Li Y, Shi X, Zhang J, Zhang X, Martin RCG (2014) Hepatic protection and anticancer activ-
ity of Curcuma: a potential chemopreventive strategy against hepatocellular carcinoma. Int
J Oncol 44:505–513
135. Thippeswamy G, Salimath BP (2006) Curcuma aromatica extract induces apoptosis and
inhibits angiogenesis in Ehrlich Ascites tumor cells in-vivo. mySCIENCE 1(1):79–92
136. Karmakar I, Dolai N, Kumar RBS, Kar B, Roy SN, Haldar PK (2013) Antitumor activity and
antioxidant property of Curcuma caesia against Ehrlich’s ascites carcinoma bearing mice.
Pharm Biol 51(6):753–759
137. Hadem KLH, Sharan RN, Kma L (2016) Phytochemicals of Aristolochia tagala and Curcuma
caesia exert anticancer effect by Tumor Necrosis Factor-α mediated decrease in Nuclear
Factor κ B binding activity. J Basic Clin Pharma 7:1–11
138. Hadisaputri YE, Miyazaki T, Suzuki S, Kubo N, Zuhrotun A (2015) Molecular characteriza-
tion of antitumor effects of the rhizome extract from Curcuma zedoaria on human esophageal
carcinoma cells. Int J Oncol 47:2255–2263
139. Khaing SL, Omar SZ, Looi CY, Arya A, Mohebali N, Mohd A (2017) Identification of active
extracts of Curcuma zedoaria and their real- time cytotoxic activities on ovarian cancer cells
and HUVEC cells. Biomed Res 28(18):9182–9187
140. Shi H, Tan B, Ji G, Lu L, Cao A, Shi S, Xie J (2013) Zedoary oil (Ezhu You) inhibits prolifera-
tion of AGS cells. Chin Med 8:13. http://www.cmjournal.org/content/8/1/13
141. Chen C-C, Chen Y, His Y-T, Chang C-S, Huang L-F, Ho C-T, Way T-D, Kao J-Y (2013)
Chemical constituents and anticancer activity of Curcuma zedoaria Roscoe essential
oil against non-small cell lung carcinoma cells in-vitro and in-vivo. J Agric Food Chem
61:11418–11427
142. Chen W, Lu Y, Gao M, Wu J, Wang A, Shi R (2011) Anti-angiogenesis effect of essential oil
from Curcuma zedoaria in-vitro and in-vivo. J Ethnopharmacol 133:220–226
143. Choi M, Kim SH, Chung W, Hwang J, Park K (2005) Xanthorrhizol, a natural sesquiterpe-
noid from Curcuma xanthorrhiza, has an anti-metastatic potential in experimental mouse
lung metastasis model. Biochem Biophys Res Commun 326:210–217
144. Cheah YH, Nordin FJ, Tee TT, Azimahtol HL, Abdullah NR, Ismail Z (2008) Antiproliferative
property and apoptotic effect of Xanthorrhizol on MDA-MB-231 breast cancer cells. Antican
Res 28:3677–3690
458 T. Soumya et al.
145. Kang Y, Park K, Chung W, Hwang J, Lee SK (2009) Xanthorrhizol, a natural sesquiterpenoid
, induces apoptosis and growth arrest in HCT116 human colon cancer cells. J Pharmacol Sci
111:276–284
146. Kim JY, An JM, Chung W, Park K, Hwang JK, Kim DS, Seo SR, Seo JT (2012) Xanthorrhizol
induces apoptosis through ROS- mediated MAPK activation in human oral squamous cell
carcinoma cells and inhibits DMBA-induced oral carcinogenesis in Hamsters. Phytother Res
27:493–498
147. Rouhollahi E, Zorofchian Moghadamtousi S, Paydar M, Fadaeinasab M, Zahedifard
M, Hajrezaie M, Abdalla Ahmed Hamdi O, Yeng Looi C, Ameen Abdulla M, Awang K,
Mohamed Z (2015) Inhibitory effect of Curcuma purpurascens BI. rhizome on HT-29 colon
cancer cells through mitochondrial-dependent apoptosis pathway. BMC Complement Altern
Med 15:15. https://doi.org/10.1186/s12906-015-0534-6
148. Hong SL, Lee GS, Syed Abdul Rahman SN, Ahmed Hamdi OA, Awang K, Aznam Nugroho
N, Abd Malek SN (2014) Essential oil content of the rhizome of Curcuma purpurascens Bl.
(Temu Tis) and its antiproliferative effect on selected human carcinoma cell lines. Sci World
J:397430. https://doi.org/10.1155/2014/397430
149. Soumya T, Lakshmipriya T, Klika KD, Jayasree PR, Manish Kumar PR (2021) Anticancer
potential of rhizome extract and a labdane diterpenoid from Curcuma mutabilis plant endemic
to Western Ghats of India. Sci Rep 11:552. https://doi.org/10.1038/s41598-020-79414-8
150. Zhang L, Yang Z, Huang Z, Zhao M, Li P, Zhou W, Zhang K, Zheng X, Lin L, Tang J, Fang
Y, Du Z (2017) Variation in essential oil and bioactive compounds of Curcuma kwangsien-
sis collected from natural habitats. Chem Biodivers 14:e1700020. https://doi.org/10.1002/
cbdv.201700020
151. Akimoto M, Iizuka M, Kanematsu R, Yoshida M, Takenaga K (2015) Anticancer effect
of Ginger extract against pancreatic cancer cells mainly through reactive oxygen species-
mediated autotic cell death. PLoS ONE 10(5):e0126605. https://doi.org/10.1371/journal.
pone.0126605
152. Qi L, Zhang Z, Zhang C, Anderson S, Liu Q, Yuan C, Wang C (2015) Anti-colon cancer
effects of 6-Shogaol through G2/M cell cycle arrest by p53/p21-cdc2/cdc25A crosstalk. Am
J Chin Med 43(4):743–756
153. Liu Q, Peng Y, Qi L, Cheng X, Xu X, Liu L, Liu E, Li P (2012) The cytotoxicity mechanism
of 6-Shogaol-treated HeLa human cervical cancer cells revealed by label-free shotgun pro-
teomics and bioinformatics analysis. Evidence-Based Complement Altern Med. https://doi.
org/10.1155/2012/278652
154. Radhakrishnan EK, Bava SV, Narayanan SS, Nath LR, Thulasidasan AKT, Soniya EV, Ruby
JA (2014) Prevents PMA-induced proliferation in colon cancer cells by inhibiting MAPK
/ AP-1 signaling. PLOS ONE 9(8):e104401. https://doi.org/10.1371/journal.pone.0104401
155. Nazim U, Jeong J, Seol J, Hur J, Eo S, Lee J, Park S (2015) Inhibition of the autophagy flux
by Gingerol enhances TRAIL-induced tumor cell death. Oncol Reports 33:2331–2336
156. Park GH, Park JH, Song HM, Eo HJ, Kim MK, Lee JW, Lee MH, Cho K, Lee JR, Cho HJ,
Jeong JB (2014) Anti-cancer activity of Ginger (Zingiber officinale) leaf through the expres-
sion of activating transcription factor 3 in human colorectal cancer cells. BMC Complement
Altern Med 14:408. http://www.biomedcentral.com/1472-6882/14/408
157. Rashid RA, Pihie AHL (2005) The antiproliferative effect of Zingiber zerumbet extracts and
fractions on the growth of human breast carcinoma cell lines. Malay J Pharm Sci 3(1):45–52
158. Rajan I, Jayasree PR, Kumar PRM (2015) Zerumbone induces mitochondria-mediated apop-
tosis via increased calcium, generation of reactive oxygen species and upregulation of soluble
histone H2AX in K562 chronic myelogenous leukemia cells. Tumor Biol 36(11):8479–8489
159. Prasannan R, Kalesh KA, Shanmugam MK, Nachiyappan A, Ramachandran L, Nguyen
AH, Prem A, Lakshmanan M, Seok K, Sethi G (2012) Key cell signaling pathways modu-
lated by Zerumbone: role in the prevention and treatment of cancer. Biochem Pharmacol
84:1268–1276
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 459
160. Koga AY, Beltrame FL, Pereira AV (2016) Several aspects of Zingiber zerumbet: a review.
Braz J Pharmacogn 26:385–391
161. Zulkhairi AM, Aspollah SM, Lian EGC, Bustamam AA (2017) Phytochemicals and cyto-
toxic studies of Zingiber cassumunar phytochemicals and cytotoxic studies of Zingiber cas-
sumunar Roxb. J Trop Agric Food Sci 45(2):187–197
162. Majdalawieh AF, Carr RI (2010) In-vitro Investigation of the potential immunomodulatory
and anti-cancer activities of Black Pepper (Piper nigrum) and Cardamom (Elettaria carda-
momum). J Med Food 13(2):371–381
163. Ray A, Jena S, Dash B, Sahoo A, Kar B, PatnaikJ PPC, Nayak S, Mahapatra N (2019)
Hedychium coronarium extract arrests cell cycle progression, induces apoptosis, and impairs
migration and invasion in HeLa cervical cancer cells. Cancer Manage Res 11:483–500
164. Endringer DC, Taveira FSN, Kondratyuk TP, Pezzuto JM, Bragaa FC (2014) Cancer chemo-
prevention activity of labdane diterpenes from rhizomes of Hedychium coronarium. Braz J
Pharmacogn 24:408–412
165. Zhana Z-J, Wena Y-T, Rena F-Y, Raob G-W, Shana W-G, Li C-P (2012) Diterpenoids and a
diarylheptanoid from Hedychium coronarium with significant anti-angiogenic and cytotoxic
activities. Chem Biodivers 9:2754–2760
166. Chen J, Hsieh M-C, Lin S, Lin C, Hsi Y-T, Lo Y-S, Chuang Y-C, Hsieh M-J, Chen MK
(2017) Coronarin D induces reactive oxygen species-mediated cell death in human naso-
pharyngeal cancer cells through inhibition of p38 MAPK and activation of JNK. Oncotarget
8(64):108006–108019
167. Lin H, Hsieh M, Hsieh Y, Yeh C, Hsueh K, Yang S (2018) Coronarin D induces apoptotic
cell death through the JNK pathway in human hepatocellular carcinoma. Environ Toxicol
33(9):946–954
168. Reddy PP, Ranga Rao R, Shashidhar J, Sastry BS, Madhusudana Rao J, Suresh Babu K
(2009) Phytochemical investigation of labdane diterpenes from the rhizomes of Hedychium
spicatum and their cytotoxic activity. Bioorg Med Chem Lett 19:6078–6081
169. Suresh G, Poornima B, Babu KS, Yadav PA, Rao MSA, Siva B, Prasad KR, Nayak VL,
Ramakrishna S (2013) Cytotoxic sesquiterpenes from Hedychium spicatum: isolation, struc-
ture elucidation and structure-activity relationship studies. Fitoterapia 86:100–107
170. Mishra T, Pal M, Meena S, Datta D, Dixit P, Kumar A, Meena B, Rana TS, Upreti DK (2016)
Composition and in-vitro cytotoxic activities of essential oil of Hedychium spicatum from
different geographical regions of western Himalaya by principal components analysis. Nat
Prod Res 30:1224–1227
171. Jing LJ, Bakar MFA, Mohamed M, Rahmat A (2011) Effects of selected Boesenbergia spe-
cies on the proliferation of several cancer cell lines. J Pharmacol Toxicol 6:272–282
172. Kirana C, Jones GP, Record IR, McIntosh GH (2007) Anticancer properties of panduratin A
isolated from Boesenbergia pandurata (Zingiberaceae). J Nat Med 61:131–137. https://doi.
org/10.1007/s11418-006-0100-0
173. Isa NM, Abdelwahab SI, Mohan S, Abdul AB, Sukari MA, Taha MME, Syam S, Narrima P,
Cheah SC, Ahmad S, Mustafa MR (2012) In vitro anti-inflammatory, cytotoxic and antioxidant
activities of boesenbergin A, a chalcone isolated from Boesenbergia rotunda (L.) (fingerroot).
Braz J Med Biol Res 45(6):524–530. https://doi.org/10.1590/S0100-879X2012007500022
174. Break MKB, Chiang M, Wiart C, Chin C-F, Khoo ASB, Khoo T-J (2020) Cytotoxic Activity of
Boesenbergiarotunda Extracts against nasopharyngeal carcinoma cells (HK1). Cardamonin,
a Boesenbergiarotunda constituent, inhibits growth and migration of HK1 cells by induc-
ing caspase-dependent apoptosis and G2/M–Phase arrest. Nutr Cancer. https://doi.org/10.108
0/01635581.2020.1751217
175. Ma XN, Xie CL, Miao Z, Yang Q, Yang XW (2017) An overview of chemical constituents
from Alpinia species in the last six decades. RSC Adv 7:14114–14144
176. Sok SPM, Arshad NM, Azmi MN, Awang K, Ozpolat B, Nagoor NH (2017) The apoptotic
effect of 1′S-1′-Acetoxychavicol Acetate (ACA) enhanced by inhibition of non-canonical
460 T. Soumya et al.
autophagy in human non-small cell lung cancer cells. PLoS One 12(2):e0171329. https://doi.
org/10.1371/journal.pone.0171329
177. Ito K, Nakazato T, Xian MJ, Yamada T, Hozumi N, Murakami A, Ohigashi H, Ikeda Y, Kizaki
M (2005) 1′-acetoxychavicol acetate is a novel nuclear factor κB inhibitor with significant
activity against multiple myeloma in vitro and in vivo. Cancer Res 65:4417–4424
178. In LLA, Arshad NM, Ibrahim H, Azmi MN, Awang K, Nagoor NH (2012) 1’-Acetoxychavicol
acetate inhibits growth of human oral carcinoma xenograft in mice and potentiates cisplatin
effect via proinflammatory microenvironment alterations. BMC Complement Altern Med
12:179. http://www.biomedcentral.com/1472-6882/12/179
179. Huang H, Chen AY, Ye X, Guan R, Rankin GO, Chen YC (2020) Galangin, a flavonoid
from lesser galangal, induced apoptosis via p53-dependent pathway in ovarian cancer cells.
Molecules 25:1579. https://doi.org/10.3390/molecules25071579
180. Kim Y, Ko H, Park J, Han I, Amor EC, Wha J, Ok H (2010) International immunopharmacol-
ogy Dimethyl cardamonin inhibits lipopolysaccharide-induced inflammatory factors through
blocking NF-κB p65 activation. Int Immunopharmacol 10:1127–1134
181. Liao Q, Shi DH, Zheng W, Xu XJ, Yu YH (2010) Antiproliferation of cardamonin is involved
in mTOR on aortic smooth muscle cells in high fructose-induced insulin resistance rats. Eur
J Pharmacol 641:179–186. https://doi.org/10.1016/j.ejphar.2010.05.024
182. Machana S, Weerapreeyakul N, Barusrux S, Nonpunya A, Sripanidkulchai B, Thitimetharoch
T (2011) Cytotoxic and apoptotic effects of six herbal plants against the human hepatocarci-
noma. Chin Med 6:39. http://www.cmjournal.org/content/6/1/39
183. Patanasethanont D, Nagai J, Matsuura C, Fukui K, Sutthanut K, Sripanidkulchai B, Yumoto
R, Takano M (2007) Modulation of function of multidrug resistance associated-proteins by
Kaempferia parviflora extracts and their components. Euro J Pharma 566:67–74
184. Ninomiya K, Chaipech TMS, Katsuyama SMY (2016) Simultaneous quantitative analysis of
12 methoxyflavones with melanogenesis inhibitory activity from the rhizomes of Kaempferia
parviflora. J Nat Med 70:179–189
185. Kuttan R, Bhanumathy P, Nirmala K, George MC (1985) Potential anticancer activity of
Turmeric (Curcuma longa). Cancer Lett 29:197–202
186. Yue GGL, Chan BCL, Hon P, Kennelly EJ, Yeung SK, Cassileth BR, Fung K, Leung P, Lau
CBS (2010) Immunostimulatory activities of polysaccharide extract isolated from Curcuma
longa. Int J Biol Macromol. 47:342–347
187. Park JH, Park KK, Kim MJ, Hwang JK, Park SK, Chung WY (2008) Cancer chemoprotective
effects of Curcuma xanthorrhiza. Phytother Res 22:695–698
188. Chung WY, Park JH, Kim MJ, Kim HO, Hwang JK, Lee SK, Park KK (2007) Xanthorrhizol
inhibits 12- O-tetradecanoylphorbol-13-acetate-induced acute inflammation and two-stage
mouse skin carcinogenesis by blocking the expression of ornithine decarboxylase , cycloox-
ygenase-2 and inducible nitric oxide synthase through mitogen-activated protein kinases and
/ or the nuclear factor- kB. Carcinogenesis 28:1224–1231
189. Oon SF, Nallappan M, Tee TT, Shohaimi S, Kassim NK, Sa’ariwijaya MSF, Cheah YH
(2015) Xanthorrhizol: a review of its pharmacological activities and anticancer properties.
Cancer Cell Int 15:100. https://doi.org/10.1186/s12935-015-0255-4
190. Xiang H, Zhang L, Xi L, Yang Y, Wang X, Lei D, Zheng X, Liu X (2018) Phytochemical
profiles and bioactivities of essential oils extracted from seven Curcuma herbs. Ind Crops
Prod 111:298–305
191. Soumya T, Jayasree PR, Deepak M, Manish Kumar PR (2019) Chemical composition, anti-
oxidant and antiproliferative activities of essential oil from rhizome and leaves of Curcuma
mutabilis Škorničk., M. Sabu & Prasanthk., endemic to Western Ghats of India. Nat Prod Res
34(16):2336–2340. https://doi.org/10.1080/14786419.2018.1533826
192. Itokawa H, Shi Q, Akiyama T, Morris-natschke SL, Lee K (2008) Recent advances in the
investigation of curcuminoids. Chin Med 3:11. https://doi.org/10.1186/1749-8546-3-11
193. Hatcher H, Planalp R, Chob J, Tortia FM, Torti SV (2008) Curcumin: from ancient medicine
to current clinical trials. Cell Mol Life Sci 65:1631–1652
Zingiberaceae Plants: A Cornucopia of Promising Chemotherapeuticals for Cancer Cure 461
194. Wilken R, Veena MS, Wang MB, Srivatsan ES (2011) Curcumin: a review of anti-cancer
properties and therapeutic activity in head and neck squamous cell carcinoma. Mol Can
10:12. http://www.molecular-cancer.com/content/10/1/12.
195. Gupta SC, Patchva S, Koh W, Aggarwal BB (2012) Discovery of Curcumin, a component
of golden spice, and its miraculous biological activities. Clini Exp Pharmacol Physiol
39:283–299
196. Amalraj A, Pius A, Sreerag G, Sreeraj G (2017) Biological activities of curcuminoids,
other biomolecules from turmeric and their derivatives - A review. J Trad Chinese Med Sci
7:205–233
197. Collett GP, Robson CN, Mathers JC, Campbell FC (2001) Curcumin modifies Apcmin apop-
tosis resistance and inhibits 2-amino 1-methyl-6-phenylimidazo [4,5-b] pyridine (PhIP)
induced tumour formation in Apcmin mice. Carcinogenesis 22:821–825
198. Hsu T-C, Young MR, Cmarik J, Colburn NH (2000) Activator protein 1 (AP-1) and Nuclear
Factor κB (NF-κB) – dependent transcriptional events in carcinogenesis. Free Radical Biol
Med 28(9):1338–1348
199. Kunnumakkara AB, Diagaradjane P, Anand P, Kuzhuvelil HB, Deorukhkar A, Gelovani J,
Guha S, Krishnan S, Aggarwal BB (2009) Curcumin sensitizes human colorectal cancer to
Capecitabine by modulation of cyclin D1, COX-2, MMP-9, VEGF and CXCR4 expression
in an orthotopic mouse model. Int J Cancer 125:2187–2197
200. Bush JA, Cheung KJ, Li G (2001) Curcumin induces apoptosis in human melanoma cells
through a Fas receptor/Caspase-8 pathway independent of p53. Exper Cell Res 271:305–314
201. Aoki H, Takada Y, Kondo S, Sawaya R, Aggarwal BB (2007) Evidence that curcumin sup-
presses the growth of malignant gliomas in-vitro and in-vivo through induction of autophagy:
role of Akt and extracellular signal-regulated kinase signaling pathways. Mol Pharmacol
72:29–39
202. Gururaj AE, Belakavadi M, Venkatesh DA, Marm D, Salimath BP (2002) Molecular mecha-
nisms of anti-angiogenic effect of Curcumin. Biochem Biophysi Res Commun 297:934–942
203. Yodkeeree S, Ampasavate C, Sung B, Aggarwal BB, Limtrakul P (2010) Demethoxycurcumin
suppresses migration and invasion of MDA-MB-231 human breast cancer cell line. Eur J
Pharmacol 627:8–15
204. Beevers CS, Chen L, Liu L, Luo Y, Webster NJG (2009) Curcumin disrupts the mammalian
target of rapamycin- raptor complex. Cancer Res 69(3):1000–1009
205. Sordillo PP, Helson L (2015) Curcumin and cancer stem cells: Curcumin has asymmetrical
effects on cancer and normal stem cells. Anticancer Res 35:599–614
206. Teiten M-H, Dicato M, Diederich M (2013) Curcumin as a regulator of epigenetic events.
Mol Nutr Food Res 57:1–11. https://doi.org/10.1002/mnfr.201300201
207. Allegra A, Innao V, Russo S, Gerace D, Alonci A, Musolino C (2016) Anticancer activity of
Curcumin and its analogues: Preclinical and clinical studies. Cancer Invest. https://doi.org/1
0.1080/07357907.2016.1247166
208. Narayanan NK, Nargi D, Randolph C, Narayanan BA (2009) Liposome encapsulation of
Curcumin and Resveratrol in combination reduces prostate cancer incidence in PTEN knock-
out mice. Int J Cancer 125:1–8
209. Liu Y, Wu Y, Yu Y, Cao C, Zhang J, Li K, Zhang P (2014) Curcumin and Resveratrol in
combination modulate drug-metabolizing enzymes as well as antioxidant indices during lung
carcinogenesis in mice. Human Exp Toxicol. https://doi.org/10.1177/0960327114551396
210. Zeng X, Cai D, Zeng Q, Chen Z, Zhong G, Zhuo J, Gan H, Huang X, Zhao Z, Yao N,
Huang D, Zhang C, Sun D, Chen Y (2017) Selective reduction in the expression of UGTs and
SULTs, a novel mechanism by which Piperine enhances the bioavailability of Curcumin in
rat. Biopharm Drug Dispos 38(1):3–19
211. Siddique RA, Harvey KA, Walker C, Altenburg J, Xu Z, Terry C, Camarillo I, Jones-hall Y,
Mariash C (2013) Characterization of synergistic anti-cancer effects of docosahexaenoic acid
and curcumin on DMBA-induced mammary tumorigenesis in mice. BMC Cancer 13:418.
http://www.biomedcentral.com/1471-2407/13/418
462 T. Soumya et al.
1 Introduction
Moringa oleifera Lam., commonly known as drumstick tree or horse radish tree, is
an important tropical medicinal plant known for its multifarious nutritional and
phytomedicinal property. It is frequently used by ethnic and local people of devel-
oped and emerging nations for their medicinal value and culinary use [1]. According
to the report of World Health Organization (WHO), nearly 70–80% of the global
population relies on herbal plants for their basic health care needs. The genus
Moringa comprises 13 species, out of which Moringa oleifera, Moringa stenopetala,
and Moringa peregrina are mostly studied because of their availability and multi-
functional properties [2]. Moringa is distributed in India, Africa, America, and
Madagascar. They are drought-resistant, fast grower (propagated through seeds/cut-
tings), and can withstand wide range of soil with minimum nutrient requirement [3].
Being a tropical deciduous tree, Moringa oleifera possesses bipinnate/tripinnate
leaves on grayish white stem with drooping branches. It also bears pendulous
25–35 cm long pods with isodiametric/ovate seeds (Fig. 1) [4].
Leaves and seeds are known to have wide range of amino acids, minerals, pro-
tein, carbohydrates, and vitamins. In addition to that, they are also known to have
important plant secondary metabolites such as polyphenols, flavonoids, moringin,
alkaloids, and tannins [5, 6]. Traditionally, Moringa was used by warriors to gain
energy and also by queens and king to maintain healthy skin and bones [7].
Medicinal, aromatic, or horticultural plants in general have nutraceutical value as
these plants are preferred as tonic or in maintaining vitality and sometimes aphro-
diasic. They are medicinally used and exploited for commercial purpose by plant-
based industries. For instance, Gymnema sylvestre is having gymnemic acid
Efforts were made to isolate and identify the potentiality of alkaloids from Moringa
leaves which indicated that they possess cardioprotective activity along with the
antihypertensive property [13].
Polyphenols are the second group of phytocompound found in both fresh and dry
leaves of Moringa conferring them antimicrobial and antioxidant properties.
Chemically, polyphenols are either in form of phenolic acid with one phenol ring or
in form of flavonoids with more than one phenol ring and they are often quantified
as tannic acid equivalent and gallic acid equivalent, respectively [12, 14, 15]. Several
researchers have identified presence of quercetin, myrecytin, and kaempferol along
with 11 more compounds through GC-MS profiling of leaves except roots, seeds,
and flowers [16, 17]. Chemical and agronomical variations were observed in acces-
sions collected and identified from India, Pakistan, and Africa which shows that
there is higher possibility of genetic variation among them regulating their meta-
bolic pathway [16, 18, 19]. Geographical and environmental variations were also
observed in some accessions of Moringa with predominant difference in tannins,
flavonoids, and polyphenols including major quantities of ellagic acid, coumaric
acid, and caffeic acid [20–22].
Another group of phytochemical known as carotenoids are coloured molecules
found in Moringa oleifera leaves as provitamin A or β-carotene which implicate
vitamin A deficiency. Carotenoids are generally characterized by their colour either
in form of yellow, red, or orange and are mostly present in vegetables and fruits
imparting their coloured complexity. Several compounds such as lutein, luteoxan-
thin, and zeaxanthin were isolated and purified from Moringa exploring their health
benefits [23]. Apart from polyphenols, flavonoids, and pigmented compounds,
Moringa leaves, pods, and seeds are known to have isothicyanate and glucosinolate
compounds. Basically, these compounds are synthesized from amino acid moieties.
A group of researchers have identified glucosinolate from wild and cultivated source
466 N. Patel and R. Krishnamurthy
Over the past years, utilization of plants with medicinal properties has increased
rapidly. Current research is focused on medicinal and nutritional property of plants
covering phytochemistry, horticulture, pharmacology, and nutraceuticals. However,
the potent phytomedicinal action often results from consortia of plant secondary
metabolites/bioactives [36]. Studies have revealed the incidence of various diseases
associated with different age groups. It was observed that the key factor responsible
for this situation is generally the weakened immune system, autoimmune diseases,
or immunosenescence [37]. Phytomedicines are termed as use of plant or plant parts
in treatment and improvement of human health. It was first coined by Henry Leclerc
in 1913. Phytomedicines are advantageous in terms of minimum side effects caused
by synthetic drugs. Moringa being a food plant has many phytomedicinal properties
(Fig. 3).
Moringa oleifera Accessions: Perspectives and Application as Nutraceuticals… 467
Moringa leaf, seeds, flowers, and roots are known to have remarkable DPPH radical
scavenging activity. This activity is conferred to any plant/ plat tissue because of
high amount of polyphenols in it. They help to scavenge reactive oxygen species
(ROS) by reducing oxidative stress. Study on antioxidant capacity of saline and
alcoholic extracts revealed that the extracts of flowers, leaf rachis, and leaves were
able to react and scavenge free radicals; however, ethanolic seed extract and saline
extracts were able to react slowly [38]. A compound myrecitin isolated from seeds
showed higher antioxidant activity when compared to BHT and α- tocopherol [39].
Free radicals are generally categorized into hydroperoxyl, hydroxyl, superoxide,
halogen, hydrogen, and nitrogen dioxide along with reactive oxygen species (ROS).
A study on methanolic extract of M. oleifera leaves depicted higher antioxidant
activity through in vitro FRAP, DPPH, and metal chelating assay [40]. Hence, the
bioactives from plants help them to stabilize/neutralize free radicals enabling them
to be used as phytomedicine.
468 N. Patel and R. Krishnamurthy
Depression is a widely known mental disorder across the globe and almost half of
the population is suffering from anxiety and mood swings. The constant state of
depression is due to oxidative stress and neurological imbalance [56]. An approach
was made to identify the efficacy of Moringa as antidepressant agent through in
vivo animal model study. It was depicted that ethanolic fraction of Moringa leaves
along with fluoxetine has potential antidepressant activity [57]. Ethanolic leaf
extract of this plant was able to minimize the chronic stress in zebrafish model [58].
Antidepressant study through tail suspension and forced swim test also indicated
the potential of n-hexane and ethanolic fraction of Moringa in relieving stress and
depression [59, 60].
Neurological disorders are also associated with nervous system mostly affecting
brain cells and spinal cord along with ganglia and nerves. Dementia (Memory loss)
mostly seen in aged people is a form of neurodegenerative disorder. Alzheimer’s
disease, Parkinson disease, Schizophrenia, and Huntington disease are associated
with reactive oxygen species and oxidative stress [61, 62]. Many efforts were made
to treat these diseases, but none of them were effective to halt their progression [63].
As the cost production of synthetic drugs is higher, natural phytocompounds were
searched and synthesized. Moringa leaf extracts (MLE) were proved to have noot-
ropic and antioxidant properties and hence several studies stated that solvent frac-
tions of Moringa can be beneficial in terms of colchicine-induced Alzheimer’s
disease in animal model study [64, 65]. Current in vitro studies using SHSY5Y
neuroblastoma cells also noted the neuroprotective effect of Moringa leaf extract
(MLE) [62].
Many studies have been undertaken to determine the role of Moringa as antimicro-
bial agent. The bioactives from Moringa leaf, stem, roots, seeds, and bark exert
potent antimicrobial property against various pathogens [66, 67]. They show inhibi-
tory activity by altering their cell permeability, growth, and multiplication rate.
Various aqueous, hexane, methanolic, and ethanolic extracts have shown potent bio-
activity against Enteropathogens, Salmonella sp., Vibrio sp., Pseudomonas sp.,
Erwinia sp., and Bacillus sp. [3, 67, 68]. Moringa roots were also found to have
antibacterial activity against peptic ulcer caused by Helicobacter pylori. It was
noted in another study that presence of pterygospermin and isothiocyanate mole-
cules in the roots attributed this phytomedicinal property to Moringa [69, 70].
Different solvent preparations of Moringa showed potent inhibitory activity against
various fungal strains such as Aspergillus sp., Fusarium sp., Alternaria sp. and
Candida sp. [71, 72]. In vitro studies of n-hexane, ethyl acetate, aqueous,
470 N. Patel and R. Krishnamurthy
study conducted on antidiabetic property of Moringa seed and aqueous leaf extract
also showed reduced level of Interleukin 6 in STZ-induced rats [85].
According to International Federation of Diabetes (IDF), about 360 million peo-
ple are affected by Diabetes mellitus (type 2) globally and it is expected to rise by
552 million by 2040 [86]. In silico study was conducted to identify the targeted
therapeutic drug that can bind the protein moiety. It was identified that anthraqui-
nones, anthocyanins, hemlock tannins, and phenolics from Moringa oleifera could
easily bind the targeted protein molecule which assisted in treatment of diabetes
mellitus [87]. Alloxan-induced diabetic rats were treated with hydroalcoholic
extract (95%) of Moringa leaves by reducing the serum glucose levels at the level of
250 mg/kg [88]. Generally, the mechanistic action of Moringa works by inhibiting
the activity of glucose transporter proteins- GLU 1 and GLU 4, thereby increasing
insulin production and treating damaged pancreatic β-cells [89].
Therapeutic potential of Moringa has been traced long time back in yielding potent
antiviral activity owing to the profound activity against HIV, EBV, HBV, HSV,
NDV, and FMDV [99–103]. The flowers, seeds, gum, root bark, and leaves were
reported to be used as immunobooster and antiviral drugs. However, evidence-based
reports were revealed to be scanty on the use of Moringa against small pox virus/
Chicken pox as world health organization has declared countries to be free from
small pox virus since May-1980 (World Health Organization, 1980) [123]. Recently,
baseline study carried out against Influenza virus depicted that Moringa A isolated
472 N. Patel and R. Krishnamurthy
from seed material was able to decrease the expression of transcription factor EB
and weaken the autophagy in virus-infected cells [104].
Skin is one of the important protective barriers and first defence system towards the
noxious pathogenic micro-organisms. As result of wound or injury, this barrier gets
disrupted which results into impairment into the connectivity of epithelial tissues. It
represents significant burden on patients affecting their mental state [105]. Generally,
the tissue regeneration/wound healing process involves hemostasis, inflammation,
proliferation, and remodelling of tissues [106]. Study on bioguided solvent fractions
of leaf was carried to ascertain the cell viability, proliferation, and wound scratch
test which depicted enhanced wound healing property. This property was known to
be attributed by vicenin-2compound isolated from methanolic fraction [107].
Recently, the intervention of nanotechnology has paved an effective way in phyto-
medicine through green synthesis. The polysaccharide extracted from Moringa seed
and its nanocomposite prepared from silver were found to be better candidate as
optimal wound dressing material [108].
4 Nutraceutical/Cosmo-Nutraceutical Value
of Moringa oleifera
The cruciferous plant Moringa oleifera (Drumstick tree) is a staple food in majority
of the countries across the globe. Due to its versatile nutritional and medicinal prop-
erty, Moringa is known as “Miracle Tree”. During ancient period, both leaves and
fruits were known to maintain skin and health of Queens and King [109]. Leaves of
this tree are worthy and precious in terms of providing nourishment to the malnour-
ished and pregnant woman. The drumstick leaves are highly packed with minerals,
vital amino acids, fatty acids, protein, and carbohydrates [110]. Most of the studies
on leaves have suggested its efficacy for combating malnutrition and also for preg-
nant women and infants [111, 112]. Moreover, Moringa oleifera is known to pro-
vide 7 × more of Vitamin C, 17 × more of Calcium, 10 × more of Vitamin A, and
25 × more of iron [113]. In emerging and underdeveloped countries where food
security is a major concern, Moringa is a great healthy diet for them. Apart from
this, immature pods are good source of fibres, minerals, and proximates [114].
As per the report suggested by Moyo and his coworkers, dietary polysaturated
fatty acid and unsaturated fatty acids were identified in dehydrated leaves of
Moringa where linolenic acid, α-linolenic acid, and linolenic acid were present in
considerate amount [115]. The culinary usage of this plant ranges from soup, salads,
to main dishes. The seed portion of this plant is well-known for highly valued ben
Moringa oleifera Accessions: Perspectives and Application as Nutraceuticals… 473
oil used as cosmeceutical, cooking, and perfume industry. The specific protein pep-
tides are also used for premature skin ageing and maintaining skin health [116]. The
inflorescence/ flowers of this miracle tree are also used in brewing infused tea and
chutney preparation due to high mineral content in them [117]. The oil separated
from Moringa seeds is also used in aromatherapy process [118]. Furthermore, the
sunscreen prepared from different herbs along with kernel oil of Moringa effec-
tively reduced UV radiation associated with conjugation system [119]. Additionally,
the facial mask prepared from leaves extracts was also demonstrated to be efficient
as cosmeceutical agent [120]. It was also observed by several researchers that the
protein and nutritional content of accessions vary from cultivar to source with sub-
stantial difference among mineral content (approx.270–271 mg/100 g Vit C;17–27%
leaf protein and 36–38% seed protein) [121, 122].
5 Conclusion
People’s accessibility to food has three dimensions including physical, social, and
pecuniary and the condition of life in the contemporary world is often alternating in
which poverty and malnutrition are the major factors. The increasing comorbidities,
malnutrition, and viral infection have become a major concern for all the age groups
and use of synthetic drug has increased the complications through their side effects.
The multipurpose phytomedicinal and nutraceutical plant Moringa could help in
maintaining food security by providing vital micronutrients, antioxidants, and pro-
tein at an economical cost. Therefore, it can be concluded that Moringa-based
phytomedicine can be a great paradigm for the future.
References
1. Anwar F, Latif S, Ashraf M, Gilani AH (2007) Moringa oleifera: a food plant with multiple
medicinal uses. Phytother Res 21(1):17–25
2. Abd Rani NZ, Husain K, Kumolosasi E (2018) Moringa genus: a review of phytochemistry
and pharmacology. Front Pharmacol 9:108. https://doi.org/10.3389/fphar.2018.00108
3. Patel N, Krishnamurthy R (2021) In-vitro Phytochemical Screening and Bioactivity of
Moringa oleifera Accessions. Biosci Biotechnol Res Commun 14(1):335–339
4. Padayachee B, Baijnath H (2012) An overview of the medicinal importance of Moringaceae.
J Med Plants Res 6(48):5831–5839
5. Rodríguez-Pérez C, Quirantes-Piné R, Fernández-Gutiérrez A, Segura-Carretero A (2015)
Optimization of extraction method to obtain a phenolic compounds-rich extract from
Moringa oleifera Lam leaves. Ind Crop Prod 66:246–254
6. Borgonovo G, De Petrocellis L, Schiano Moriello A, Bertoli S, Leone A, Battezzati A,
Mazzini S, Bassoli A (2020) Moringin, a stable Isothiocyanate from Moringa oleifera, acti-
vates the somatosensory and pain receptor TRPA1 channel in vitro. Molecules 25(4):976
7. Mahmood KT, Mugal T, Haq IU (2010) Moringa oleifera: a natural gift-A review. J Pharm
Sci Res 2(11):775–781
474 N. Patel and R. Krishnamurthy
8. Krishnamurthy R, Chandorkar MS, Pathak JM, Animasaun DA, Gupta R (2015) Selection
of elite lines from accessions of Gymnema sylvestre (Gudmar) based on characterization of
foliage and gymnemic acid yield. Int J Med Plants Photon 108:596–605
9. Krishnamurthy R, Animasaun DA, Patel RT, Ingalhalli RS (2016) Phytochemical constitu-
ents and hypoglycemic effect of gymnemic acid extracts from big and small leaf varieties of
Gymnema sylvestre R. Br. Indonesian J Pharm 27(2):59
10. Kim HS (2005) Do not put too much value on conventional medicines. J Ethnopharmacol
100(1–2):37–39
11. Bohn T, Blackwood M, Francis D, Tian Q, Schwartz SJ, Clinton SK (2013) Bioavailability of
phytochemical constituents from a novel soy fortified lycopene rich tomato juice developed
for targeted cancer prevention trials. Nutr Cancer 65(6):919–929
12. Ma ZF, Ahmad J, Zhang H, Khan I, Muhammad S (2020) Evaluation of phytochemical and
medicinal properties of Moringa (Moringa oleifera) as a potential functional food. S Afr J
Bot 129:40–46
13. Dangi SY, Jolly CI, Narayanan S (2002) Antihypertensive activity of the total alkaloids from
the leaves of Moringa oleifera. Pharm Biol 40(2):144–148
14. Tesfay SZ, Magwaza LS, Mbili N, Mditshwa A (2017) Carboxyl methylcellulose (CMC)
containing Moringa plant extracts as new postharvest organic edible coating for Avocado
(Persea americana Mill.) fruit. Sci Hortic 226:201–207
15. Kumar S, Pandey AK (2013) Chemistry and biological activities of flavonoids: an overview.
Sci World J 2013:1–16
16. Saini RK, Sivanesan I, Keum YS (2016) Phytochemicals of Moringa oleifera: a review of
their nutritional, therapeutic and industrial significance. 3 Biotech 6(2):1–14
17. Igwe KK, Nwankwo PO, Otuokere IE, Ijioma S, Amaku F (2015) GCMS analysis of phy-
tocomponents in the methanolic extract of Moringa oleifera leave. Int J Res Pharm Sci
2(11):1–6
18. Rajalakshmi R, Rajalakshmi S, Parida A (2017) Evaluation of the genetic diversity and
population structure in drumstick (Moringa oleifera L.) using SSR markers. Curr Sci
112:1250–1256
19. Popoola J, Igwe D, Jegede O, Iwu V, Adegbite A, Omonhinmin C (2019) Agronomic prac-
tices, genetic diversity and population structure of Moringa oleifera (Lam.) in Nigeria. J Adv
Res Dynam Control Syst 12:659–670
20. Vongsak B, Sithisarn P, Gritsanapan W (2013) Simultaneous determination of crypto-
chlorogenic acid, isoquercetin, and astragalin contents in Moringa oleifera leaf extracts by
TLC-densitometric method. Evid-Based Complement Alternat Med 2013
21. Oboh G, Ademiluyi AO, Ademosun AO, Olasehinde TA, Oyeleye SI, Boligon AA, Athayde
ML (2015) Phenolic extract from Moringa oleifera leaves inhibits key enzymes linked to
erectile dysfunction and oxidative stress in rats’ penile tissues. Biochem Res Int 2015
22. Leone A, Spada A, Battezzati A, Schiraldi A, Aristil J, Bertoli S (2015) Cultivation, genetic,
ethnopharmacology, phytochemistry and pharmacology of Moringa oleifera leaves: an over-
view. Int J Mol Sci 16(6):12791–12835
23. Saini RK, Prashanth KH, Shetty NP, Giridhar P (2014) Elicitors, SA and MJ enhance carot-
enoids and tocopherol biosynthesis and expression of antioxidant related genes in Moringa
oleifera Lam. leaves. Acta Physiol Plant 36(10):2695–2704
24. Chodur GM, Olson ME, Wade KL, Stephenson KK, Nouman W, Fahey JW (2018) Wild
and domesticated Moringa oleifera differ in taste, glucosinolate composition, and antioxidant
potential, but not myrosinase activity or protein content. Sci Rep 8(1):1–10
25. Amaglo NK, Bennett RN, Curto RBL, Rosa EA, Turco VL, Giuffrida A et al (2010) Profiling
selected phytochemicals and nutrients in different tissues of the multipurpose tree Moringa
oleifera L., grown in Ghana. Food Chem 122(4):1047–1054
26. Nair A, James TJ (2020) Computational screening of phytocompounds from Moringa oleif-
era leaf as potential inhibitors of SARS-CoV-2 Mpro
Moringa oleifera Accessions: Perspectives and Application as Nutraceuticals… 475
27. Bhattacharya A, Naik MR, Agrawal D, Rath K, Kumar S, Mishra SS (2014) Antipyretic, anti-
inflammatory and analgesic effects of leaf extract of drumstick tree. J Young Pharm 6(4):20
28. Saini RK, Manoj P, Shetty NP, Srinivasan K, Giridhar P (2014) Dietary iron supplements
and Moringa oleifera leaves influence the liver hepcidin messenger RNA expression and
biochemical indices of iron status in rats. Nutr Res 34(7):630–638
29. Mensah JK, Ikhajiagbe B, Edema NE, Emokhor J (2012) Phytochemical, nutritional and
antibacterial properties of dried leaf powder of Moringa oleifera (Lam.) from Edo Central
Province, Nigeria. J Nat Prod Plant Resour 2(1):107–112
30. Bhattacharya A, Tiwari P, Sahu PK, Kumar S (2018) A review of the phytochemical and phar-
macological characteristics of Moringa oleifera. J Pharm Bioallied Sci 10(4):181
31. Du Toit ES, Sithole J, Vorster J (2020) Leaf harvesting severity affects total phenolic and tan-
nin content of fresh and dry leaves of Moringa oleifera Lam. trees growing in Gauteng, South
Africa. S Afr J Bot 129:336–340
32. Tian X, Tang H, Lin H, Cheng G, Wang S, Zhang X (2013) Saponins: the potential che-
motherapeutic agents in pursuing new anti-glioblastoma drugs. Mini Rev Med Chem
13(12):1709–1724
33. Popoola JO, Obembe OO (2013) Local knowledge, use pattern and geographical distribution
of Moringa oleifera Lam.(Moringaceae) in Nigeria. J Ethnopharmacol 150(2):682–691
34. Balakrishnan V, Prema P, Ravindran KC, Robinson JP (2009) Ethnobotanical studies among
villagers from Dharapuram taluk, Tamil Nadu, India. Glob J Pharmacol 3(1):08–14
35. Gandji K, Chadare FJ, Idohou R, Salako VK, Assogbadjo AE, Kakaï RG (2018) Status and
utilisation of Moringa oleifera Lam: a review. Afr Crop Sci J 26(1):137–156
36. Briskin DP (2000) Medicinal plants and phytomedicines. Linking plant biochemistry and
physiology to human health. Plant Physiol 124(2):507–514
37. Hong H, Wang Q, Li J, Liu H, Meng X, Zhang H (2019) Aging, cancer and immunity. J
Cancer 10(13):3021
38. Santos AF, Argolo AC, Paiva PM, Coelho LC (2012) Antioxidant activity of Moringa oleifera
tissue extracts. Phytother Res 26(9):1366–1370
39. Lalas S, Tsaknis J (2002) Extraction and identification of natural antioxidant from the seeds
of the Moringa oleifera tree variety of Malawi. J Am Oil Chem Soc 79(7):677–683
40. Mahmoud KB, Wasli H, Mansour RB, Jemai N, Selmi S, Jemmali A, Ksouri R (2022)
Antidiabetic, antioxidant and chemical functionalities of Ziziphus jujuba (Mill.) and Moringa
oleifera (Lam.) plants using multivariate data treatment. S Afr J Bot 144:219–228
41. Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL Jr, Daniel WJ, Barry
JM, Suzzane O, Jackson TW, Edward JR, National High Blood Pressure Education Program
Coordinating Committee (2003) Seventh report of the joint national committee on prevention,
detection, evaluation, and treatment of high blood pressure. Hypertension 42(6):1206–1252
42. Acuram LK, Chichioco Hernandez CL (2019) Anti-hypertensive effect of Moringa oleifera
Lam. Cogent Biol 5(1):1596526
43. Adefegha SA, Oboh G, Iyoha AE, Oyagbemi AA (2019) Comparative effects of horserad-
ish (Moringa oleifera) leaves and seeds on blood pressure and crucial enzymes relevant to
hypertension in rat. Pharma Nutr 9:100–152
44. Singh BN, Singh BR, Singh RL, Prakash D, Dhakarey R, Upadhyay G, Singh HB (2009)
Oxidative DNA damage protective activity, antioxidant and anti-quorum sensing potentials
of Moringa oleifera. Food Chem Toxicol 47(6):1109–1116
45. Hammam MA, Kalil GA, El-Sayed SM, Ahmed IA (2016) Effects of Moringa oleifera Lam
(Moringaceae) seeds in rats fed with high-fat diet. J Pharm Chem Biol Sci 4(1):76–87
46. Randriamboavonjy JI, Rio M, Pacaud P, Loirand G, Tesse A (2017) Moringa oleifera seeds
attenuate vascular oxidative and nitrosative stresses in spontaneously hypertensive rats.
Oxidative Med Cell Longev 2017
47. Rauf A, Akram M, Semwal P, Mujawah AA, Muhammad N, Riaz Z, Khan H (2021)
Antispasmodic potential of medicinal plants: a comprehensive review. Oxid Med Cell
Longev 2021
476 N. Patel and R. Krishnamurthy
48. Cáceres A, Saravia A, Rizzo S, Zabala L, De Leon E, Nave F (1992) Pharmacologie proper-
ties of Moringa oleifera. 2: screening for antispasmodic, anti inflammatory and diuretic activ-
ity. J Ethnopharmacol 36(3):233–237
49. Gilani AH, Aftab K, Suria A, Siddiqui S, Salem R, Siddiqui BS, Faizi S (1994) Pharmacological
studies on hypotensive and spasmolytic activities of pure compounds from Moringa oleifera.
Phytother Res 8(2):87–91
50. El-bakry K, Toson ES, Serag M, Aboser M (2016) Hepatoprotective effect of Moringa oleif-
era leaves extract against carbon tetrachloride-induced liver damage in rats. World J Pharm
Pharm Sci 5:76–89
51. Elhamalawy OH, Al-Anany FS, El Makawy AI (2022) Thiamethoxam-induced hematologi-
cal, biochemical, and genetic alterations and the ameliorated effect of Moringa oleifera in
male mice. Toxicology Reports
52. Fakurazi S, Hairuszah I, Nanthini U (2008) Moringa oleifera Lam prevents acetamino-
phen induced liver injury through restoration of glutathione level. Food Chem Toxicol
46(8):2611–2615
53. Ouédraogo M, Lamien-Sanou A, Ramdé N, Ouédraogo AS, Ouédraogo M, Zongo SP,
Guissou PI (2013) Protective effect of Moringa oleifera leaves against gentamicin-induced
nephrotoxicity in rabbits. Exp Toxicol Pathol 65(3):335–339
54. Aristianti A, Nurkhaeri N, Tandiarrang VY, Awaluddin A, Muslimin L (2021) Formulation
and pharmacological studies of leaves of Moringa (Moringa oleifera), a novel hepatoprotec-
tion in oral drug formulations. Open Access Macedonian J Med Sci 9(A):151–156
55. Singh SK, Rajoria K, Kushal A, Dadhich S (2021) Moringa oleifera lam. a drug with
ayurvedic and biomedicine approaches. J Ayurveda 15(4):293
56. Pedersen ME, Szewczyk B, Stachowicz K, Wieronska J, Andersen J, Stafford GI et al
(2008) Effects of South African traditional medicine in animal models for depression. J
Ethnopharmacol 119(3):542–548
57. Kaur G, Invally M, Sanzagiri R, Buttar HS (2015) Evaluation of the antidepressant activity of
Moringa oleifera alone and in combination with fluoxetine. J Ayurveda Integr Med 6(4):273
58. Rosdy MS, Rofiee MS, Samsulrizal N, Salleh MZ, Kek TL (2021) Understanding the effects
of Moringa oleifera in chronic unpredictable stressed zebrafish using metabolomics analysis.
J Ethnopharmacol:114290
59. Yunusa S, Kura AU, Ladan AA, Magaji SY (2018) Preliminary phytochemical analysis and
antidepressant activity of n-hexane fraction of Moringa oleifera ethanol leaf extract in mice.
Acta Sci Pharm Sci 2:84–88
60. Yadav J, Satish KS, Lalit S (2016) Evaluation of antidepressant activity of leaves extract of
Moringa oliefera by using FST and TST model on Swiss Albino Mice. World J Pharm Res
5:967–976
61. Feng C, Luo T, Zhang S, Liu K, Zhang Y, Luo Y, Ge P (2016) Lycopene protects human
SH-SY5Y neuroblastoma cells against hydrogen peroxide-induced death via inhibi-
tion of oxidative stress and mitochondria-associated apoptotic pathways. Mol Med Rep
13(5):4205–4214
62. Hashim JF, Vichitphan S, Boonsiri P, Vichitphan K (2021) Neuroprotective assessment of
Moringa oleifera leaves extract against oxidative-stress-induced cytotoxicity in SHSY5Y
neuroblastoma cells. Plan Theory 10(5):889
63. Kou X, Chen N (2017) Resveratrol as a natural autophagy regulator for prevention and treat-
ment of Alzheimer’s disease. Nutrients 9(9):927
64. Ganguly R, Guha D (2008) Alteration of brain monoamines & EEG wave pattern in rat model
of Alzheimer’s disease & protection by Moringa oleifera. Indian J Med Res 128(6)
65. Ghimire S, Subedi L, Acharya N, Gaire BP (2021) Moringa oleifera: a tree of life as a prom-
ising medicinal plant for neurodegenerative diseases. J Agric Food Chem 69:14358–14371
66. Kou X, Li B, Olayanju JB, Drake JM, Chen N (2018) Nutraceutical or pharmacological
potential of Moringa oleifera Lam. Nutrients 10(3):343
Moringa oleifera Accessions: Perspectives and Application as Nutraceuticals… 477
67. Prabakaran M, Kim SH, Sasireka A, Chandrasekaran M, Chung IM (2018) Polyphenol com-
position and antimicrobial activity of various solvent extracts from different plant parts of
Moringa oleifera. Food Biosci 26:23–29
68. Rahman MM, Rahman MM, Akhter S, Jamal MA, Pandeya DR, Haque MA et al (2010)
Control of coliform bacteria detected from diarrhea associated patients by extracts of
Moringa oleifera. Nepal Med Coll J 12(1):12–19
69. Farooq F, Rai M, Tiwari A, Khan AA, Farooq S (2012) Medicinal properties of Moringa
oleifera: an overview of promising healer. J Med Plants Res 6(27):4368–4374
70. Mishra G, Singh P, Verma R, Kumar S, Srivastav S, Jha KK, Khosa RL (2011) Traditional
uses, phytochemistry and pharmacological properties of Moringa oleifera plant: an overview.
Pharm Lett 3(2):141–164
71. Aondo TO, Odiaka NI, Akesa TM, Olaleye OO (2018) Phytochemical and antifungal effi-
cacy of different parts of Moringa oleifera plant extracts. Asian J Biotechnol Bioresour
Technol:1–8
72. Patel N, Mohan JSS (2018) Antimicrobial activity and phytochemical analysis of Moringa
oleifera Lam. crude extracts against selected bacterial and fungal strains. Int J Pharmacogn
Phytochem Res 10(02):68–79
73. Oniha M, Eni A, Akinnola O, Omonigbehin EA, Ahuekwe EF, Olorunshola JF (2021) In vitro
antifungal activity of extracts of Moringa oleifera on phytopathogenic fungi affecting Carica
papaya. Open Access Macedonian J Med Sci 9(A):1081–1085
74. Kamuhabwa A, Nshimo C, de Witte P (2000) Cytotoxicity of some medicinal plant extracts
used in Tanzanian traditional medicine. J Ethnopharmacol 70(2):143–149
75. Jung IL (2014) Soluble extract from Moringa oleifera leaves with a new anticancer activity.
PLoS One 9(4):e95492
76. Purwal L, Pathak AK, Jain UK (2010) In vivo anticancer activity of the leaves and fruits of
Moringa oleifera on mouse melanoma. Pharmacol Online 1:655–665
77. Sreelatha S, Jeyachitra A, Padma PR (2011) Antiproliferation and induction of apoptosis by
Moringa oleifera leaf extract on human cancer cells. Food Chem Toxicol 49(6):1270–1275
78. Khalafalla MM, Abdellatef E, Dafalla HM, Nassrallah AA, Aboul-Enein KM, Lightfoot DA
et al (2010) Active principle from Moringa oleifera Lam leaves effective against two leuke-
mias and a hepatocarcinoma. Afr J Biotechnol 9(49):8467–8471
79. Kiran MS, Kumar CR, Shwetha UR, Onkarappa HS, Betageri VS, Latha MS (2021) Green
synthesis and characterization of gold nanoparticles from Moringa oleifera leaves and assess-
ment of antioxidant, antidiabetic and anticancer properties. Chem Data Collect 33:100714
80. Aboulthana WM, Shousha WG, Essawy EAR, Saleh MH, Salama AH (2021) Assessment of
the anti-cancer efficiency of silver Moringa oleifera leaves nano-extract against colon cancer
induced chemically in rats. Asian Pac J Cancer Prev 22(10):3267–3286
81. Schnell O, Standi E (2006) Impaired glucose tolerance, diabetes, and cardiovascular disease.
Endocr Pract 12:16–19
82. Edoga CO, Njoku OO, Amadi EN, Okeke JJ (2013) Blood sugar lowering effect of Moringa
oleifera Lam. in albino rats. Int J Sci Technol 3(1):88–90
83. Rana TS, Singh KK, Rao RR (2000) Studies on indigenous herbal remedies for diabetes
mellitus in India. In: Ethnobotany and medicinal plants of Indian subcontinent. Scientific
Publishers Jodhpur, p 115
84. Yassa HD, Tohamy AF (2014) Extract of Moringa oleifera leaves ameliorates streptozotocin-
induced diabetes mellitus in adult rats. Acta Histochem 116(5):844–854
85. Al-Malki AL, El Rabey HA (2015, 2015) The antidiabetic effect of low doses of Moringa
oleifera Lam. seeds on streptozotocin induced diabetes and diabetic nephropathy in male rats.
BioMed Res Int
86. Ogurtsova K, da Rocha Fernandes JD, Huang Y, Linnenkamp U, Guariguata L, Cho NH,
Makaroff LE (2017) IDF Diabetes Atlas: global estimates for the prevalence of diabetes for
2015 and 2040. Diabetes Res Clin Pract 128:40–50
478 N. Patel and R. Krishnamurthy
87. Zainab B, Ayaz Z, Alwahibi MS, Khan S, Rizwana H, Soliman DW, Abbasi AM (2020)
In-silico elucidation of Moringa oleifera phytochemicals against diabetes mellitus. Saudi J
Biol Sci 27(9):2299–2307
88. Kar A, Choudhary BK, Bandyopadhyay NG (2003) Comparative evaluation of hypogly-
caemic activity of some Indian medicinal plants in alloxan diabetic rats. J Ethnopharmacol
84(1):105–108
89. Pradana DLC, Rahmi EP, Muti AF (2021) Hypoglycemic effect of Moringa oleifera aqueous
extract in diabetic animal studies: a mechanisms review. Diabetes 57:6
90. Agrawal B, Mehta A (2008) Antiasthmatic activity of Moringa oleifera Lam.: a clinical
study. Indian J Pharmacol 40(1):28
91. Shifren A, Witt C, Christie C, Castro M (2012) Mechanisms of remodeling in asthmatic
airways. J Allergy 2012
92. Mahajan SG, Banerjee A, Chauhan BF, Padh H, Nivsarkar M, Mehta AA (2009) Inhibitory
effect of n-butanol fraction of Moringa oleifera Lam. seeds on ovalbumin-induced airway
inflammation in a Guinea pig model of asthma. Int J Toxicol 28(6):519–527
93. Bartosch R, Feldberg W, Nagel E (1932) Release of a histamine-like substance in Guinea pig
anaphylaxis. Pfluger’s Arch Complete Physiol Man Anim 230(1):129–153
94. Irie M, Nagata S, Endo Y (2002) Effect of isolation on classical conditioned histamine release
in Guinea pigs. Neurosci Res 44(1):31–35
95. Li RR, Pang LL, Du Q, Shi Y, Dai WJ, Yin KS (2010) Apigenin inhibits allergen-induced
airway inflammation and switches immune response in a murine model of asthma.
Immunopharmacol Immunotoxicol 32(3):364–370
96. Wang J, Fang X, Ge L, Cao F, Zhao L, Wang Z, Xiao W (2018) Antitumor, antioxidant and
anti-inflammatory activities of kaempferol and its corresponding glycosides and the enzy-
matic preparation of kaempferol. PLoS One 13(5):e0197563
97. Suresh S, Chhipa AS, Gupta M, Lalotra S, Sisodia SS, Baksi R, Nivsarkar M (2020)
Phytochemical analysis and pharmacological evaluation of methanolic leaf extract of
Moringa oleifera lam. In ovalbumin induced allergic asthma. S Afr J Bot 130:484–493
98. Nworu CS, Okoye EL, Ezeifeka GO, Esimone CO (2013) Extracts of Moringa oleifera Lam.
showing inhibitory activity against early steps in the infectivity of HIV-1 lentiviral particles
in a viral vector-based screening. Afr J Biotechnol 12(30)
99. Murakami A, Kitazono Y, Jiwajinda S, Koshimizu K, Ohigashi H (1998) Niaziminin, a thio-
carbamate from the leaves of Moringa oleifera, holds a strict structural requirement for inhi-
bition of tumor-promoter-induced Epstein-Barr virus activation. Planta Med 64(04):319–323
100. Imran I, Altaf I, Ashraf M, Javeed A, Munir N, Bashir R (2016) In vitro evaluation of antiviral
activity of leaf extracts of Azadirachta indica, Moringa oleifera, and Morus alba against the
foot and mouth disease virus on BHK-21 cell line. Sci Asia 42(6):392–396
101. Eze DC, Okwor EC, Okoye JO, Onah DN (2013) Immunologic effects of Moringa oleifera
methanolic leaf extract in chickens infected with Newcastle disease virus (kudu 113) strain.
Afr J Pharm Pharmacol 7(31):2231–2237
102. Feustel S, Ayón-Pérez F, Sandoval-Rodriguez A, Rodríguez-Echevarría R, Contreras-Salinas
H, Armendáriz-Borunda J, Sánchez-Orozco LV (2017, 2017) Protective effects of Moringa
oleifera on HBV genotypes C and H transiently transfected Huh7 cells. J Immunol Res
103. Xiong Y, Rajoka MSR, Mehwish HM, Zhang M, Liang N, Li C, He Z (2021) Virucidal activ-
ity of Moringa A from Moringa oleifera seeds against Influenza A Viruses by regulating
TFEB. Int Immunopharmacol 95:107561
104. Nagori BP, Solanki R (2011) Role of medicinal plants in wound healing. Res J Med Plant
5(4):392–405
105. Guo SA, DiPietro LA (2010) Factors affecting wound healing. J Dent Res 89(3):219–229
106. Muhammad AA, Pauzi NAS, Arulselvan P, Abas F, Fakurazi S (2013) In vitro wound healing
potential and identification of bioactive compounds from Moringa oleifera Lam. BioMed
Res Int 2013
Moringa oleifera Accessions: Perspectives and Application as Nutraceuticals… 479
107. Mehwish HM, Liu G, Rajoka MSR, Cai H, Zhong J, Song X, He Z (2021) Therapeutic
potential of Moringa oleifera seed polysaccharide embedded silver nanoparticles in wound
healing. Int J Biol Macromol 184:144–158
108. Sujatha BK, Patel P (2017) Moringa oleifera–Nature’s Gold. Imp J Interdiscip Res
3(5):1175–1179
109. Rathnayake ARMHA, Navaratne SB, Uthpala TG (2019) Moringa olifera plant and the nutri-
tional and medicinal properties of Moringa olifera leaves. In: Trends & prospects in process-
ing of horticultural crops, pp. 251–268
110. Okiki PA, Osibote IA, Balogun O, Oyinloye BE, Idris OO, Adelegan O, Olagbemide PT
(2015) Evaluation of proximate, minerals, vitamins and phytochemical composition of
Moringa oleifera Lam. cultivated in Ado Ekiti, Nigeria. Adv Biol Res 9(6):436–443
111. Mishra SP, Singh P, Singh S (2012) Processing of Moringa oleifera leaves for human con-
sumption. Bull Environ Pharmacol Life Sci 2(1):28–31
112. Rockwood JL, Anderson BG, Casamatta DA (2013) Potential uses of Moringa oleifera and
an examination of antibiotic efficacy conferred by M. oleifera seed and leaf extracts using
crude extraction techniques available to underserved indigenous populations. Int J Phytother
Res 3(2):61–71
113. Mahato DK, Kargwal R, Kamle M, Sharma B, Pandhi S, Mishra S et al (2022)
Ethnopharmacological properties and nutraceutical potential of Moringa oleifera. Phytomed
Plus 2(1):100168
114. Moyo B, Masika PJ, Hugo A, Muchenje V (2011) Nutritional characterization of Moringa
(Moringa oleifera Lam.) leaves. Afr J Biotechnol 10(60):12925–12933
115. Sandeep G, Anitha T, Vijayalatha KR, Sadasakthi A (2019) Moringa for nutritional security
(Moringa oleifera Lam.). Int J Bot Stud 4:21–24
116. Brilhante RSN, Sales JA, Pereira VS, Castelo DDSCM, de Aguiar Cordeiro R, de Souza
Sampaio CM et al (2017) Research advances on the multiple uses of Moringa oleifera: a sus-
tainable alternative for socially neglected population. Asian Pac J Trop Med 10(7):621–630
117. Armand-Stussi I, Basocak V, Pauly G, McCaulley J (2003) Moringa oleifera: an interesting
source of active ingredients for skin and hair care. SÖFW-J 129(9):45–52
118. Kale S, Gaikwad M, Bhandare S (2011) Determination and comparison of in vitro SPF of
topical formulation containing Lutein ester from Tagetes erecta L Flowers, Moringa oleifera
Lam seed oil and Moringa oleifera Lam seed oil containing lutein ester. Int J Res Pharm
Biomed Sci 2(3):1220–1224
119. Hendrawati H, Azizah YN, Hapsari NK (2021) Facial mask formulation enriched with
Moringa leaves (Moringa oleifera) extract and their activity as antioxidants and Antibacterials.
J Kimia Valensi 6(2):198–207
120. Jongrungruangchok S, Bunrathep S, Songsak T (2010) Nutrients and minerals con-
tent of eleven different samples of Moringa oleifera cultivated in Thailand. J Health Res
24(3):123–127
121. Saini RK, Shetty NP, Prakash M, Giridhar P (2014) Effect of dehydration methods on reten-
tion of carotenoids, tocopherols, ascorbic acid and antioxidant activity in Moringa oleifera
leaves and preparation of a RTE product. J Food Sci Technol 51(9):2176–2182
122. Goswami D, Mukherjee PK, Kar A, Ojha D, Roy S, Chattopadhyay D (2016) Screening
of ethnomedicinal plants of diverse culture for antiviral potentials. Indian J Tradit Knowl
15:474–481
123. World Health Organization (1980) The global eradication of smallpox: final report of the
Global Commission for the Certification of Smallpox Eradication, Geneva, December 1979.
World Health Organization
An Overview of Ethnobotany,
Phytochemicals, and Pharmacological
Properties of Ficus Species
1 Introduction
Nature has been a source of medicinal agents for thousands of years and an impres-
sive number of modern drugs have been derived from natural sources; many of these
isolations were based on the uses of the agents in traditional medicine [1]. The
World Health Organization (WHO) defines traditional medicine as the “diverse
health practices, approaches, knowledge and beliefs incorporating plant, animal-
and/or mineral-based medicines, spiritual therapies, manual techniques and exer-
cises applied singularly or in combination to maintain well-being, as well as to treat,
diagnose, or prevent illness”. It is clear, however, that there is a need to validate the
information through an organized infrastructure for it to be used as an effective
therapeutic means, either in conjunction with existing therapies, or as a tool in novel
drug discovery. It is therefore imperative to study medicinal plants and to determine
and evaluate their potential biological activities and efficacy. The use of natural
S. P. Sasidharan
Department of Botany, NSS College Nemmara, Palakkad, Kerala, India
X. Yang
Key Laboratory of Economic Plants and Biotechnology and the Yunnan Key Laboratory for
Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences,
Kunming, China
Center for Studies in Stem Cells, Cell Therapy and Toxicological Genetics (CeTroGen),
Faculty of Medicine, Federal University of Mato Grosso do Sul,
Campo Grande, Mato Grosso do Sul, Brazil
K. Arunachalam (*)
Center for Studies in Stem Cells, Cell Therapy and Toxicological Genetics (CeTroGen),
Faculty of Medicine, Federal University of Mato Grosso do Sul,
Campo Grande, Mato Grosso do Sul, Brazil
wild or cultivated plants. Natural compounds can thus be lead compounds, allowing
the design, development, and the discovery of new therapeutic agents [8].
A search in the natural product alert database suggested that only about 15% of
all plant species had been studied to some extent for their phytochemistry and only
about 5% for one or more biological activities [9]. Although extensive research on
medicinal plants is published every year, only a few plants have been comprehen-
sively studied for their pharmacological properties. Thus, traditional medicines and
medicinal plants obviously represent a great source of novel medicines and leads for
drug development.
Living organisms require ample amount of oxygen for their metabolism and
energy production. However, free radicals are produced during the energy produc-
tion process [10], as the unavoidable consequence of respiration in aerobic organ-
isms. Free radicals have very important role in origin of life and biological evolution,
leaving beneficial effects on the organisms [11]. Free radicals are atomic or molecu-
lar species with unpaired electrons in the outermost bonding orbital and are likely
to take part in chemical reactions. Electrons prefer to be in pairs and when an elec-
tron is alone in its orbital, it will try to take an electron from another atom to become
more stable. When the other atom loses its electron, it tries on its turn to steal an
electron from another atom, often resulting in a dangerous chain reaction. Free radi-
cal can cause damage to our cells, but they also play an important role in a number
of biological processes, such as the intracellular killing of bacteria by the white
blood cells and some cell signaling processes. Overproduction of free radicals can
cause oxidative damage to biomolecules (lipids, proteins, and DNA), eventually
leading to many chronic diseases such as atherosclerosis, cancer, diabetics, rheuma-
toid arthritis, postischemic perfusion injury, myocardial infarction, cardiovascular
diseases, chronic inflammation, stroke and septic shock, aging, and other degenera-
tive diseases in humans [12, 13].
Pain and inflammation are associated with virtually all diseases. Typical inflam-
matory diseases such as rheumatoid arthritis, asthma, colitis, and hepatitis are the
leading cause of disability and death [14]. Recently, chronic inflammation has been
found to contribute to the development of cancer, cardiovascular, and neurodegen-
erative diseases [15]. Analgesic and anti-inflammatory 4 drugs relieve mild to mod-
erate pain and reduce inflammation. All these analgesic and anti-inflammatory
agents produce their therapeutic effects by inhibiting various prostaglandins sub-
stances involved in development of pain and inflammation as well as regulation of
body temperature. Due to extensive use of analgesic and anti-inflammatory agents,
the toxicity and untoward effects do occur many times especially when therapy of
pain and inflammation involves use of higher dose for longer period [16].
The conventional drugs used for the management of pain and inflammation
include the steroidal and nonsteroidal anti-inflammatory agents as well as the opi-
ates. It is estimated that about 30 million people worldwide take nonsteroidal anti-
inflammatory drugs [17, 18]. However, their prolonged use has been associated with
serious side effects such as gastric ulceration, hemorrhage, bronchospasm, and kid-
ney dysfunction [19]. Also, because of adverse effects of such dyslipidaemia,
Cushing’s syndrome, hypertension and immunosuppression by steroidal
484 S. P. Sasidharan et al.
anti-inflammatory drugs and tolerance and dependence induced by opiates, the use
of these drugs as anti-inflammatory and analgesic agents has not been successful in
some cases [20]. Therefore, there has been an intense search for new anti-
inflammatory and analgesic drugs lacking these side effects as alternatives to these
drugs. In this direction, attention has been focused on the investigation of the effi-
cacy of plant-based drugs used in traditional medicine. This is because investigation
of the efficacy of certain plant-based drugs used in traditional medicine led to the
discovery of potent anti-inflammatory agents in clinical use such as aspirin and
colchicine [21].
Other natural products, with marked anti-inflammatory effects, which have pro-
vided effective adjuncts to the management of inflammatory conditions include:
omega 3-fatty acids from fish oil, curcumin from the spice turmeric (Curcuma
longa), frankincense from Boswellia serrata, capsaicin from Capsicum annum, and
ginger root extract from Zingiber officinalis [22]. These agents have been used suc-
cessfully as adjuncts in several chronic inflammatory diseases [23]. Thus, plants
still present a large source of compounds that might serve as leads for the develop-
ment of novel anti-inflammatory drugs. Unlike their synthetic counterparts, they
have not been shown to accelerate cartilage destruction or produce 5 liver and kid-
ney toxicities [24]. For these reasons, natural products can be considered as viable
alternatives to conventional anti-inflammatory drugs in a large percentage of patients
suffering from various inflammatory diseases.
The excessive production of reactive oxygen metabolites by phagocytic leuco-
cytes during the inflammatory process, as part of host defence, deregulates cellular
function causing tissue injury which in turn augments the state of inflammation
leading to chronic inflammatory diseases [25]. These oxidants also inhibit wound
healing. Antioxidants, which scavenge these reactive oxygen metabolites, have
been found to complement the anti-inflammatory process, promoting tissue repair
and wound healing. A number of plant secondary metabolites such as apigenin,
quercetin, luteolin, and silymarin have been found to exhibit anti-inflammatory
activities due to their antioxidant properties [26, 27]. Thus, antioxidative phyto-
chemicals, especially phenolic compounds, found in vegetables, fruits, and medici-
nal plants could be explored for their potential role in the prevention of inflammatory
diseases.
Plants have also been a source of various anti-infective agents. The bacteriostatic
and fungicidal properties of lichens and the antimicrobial action of allicin in garlic
(Allium sativum) are a few examples of age-old antibacterial therapy [28]. An
expansive range of plants belonging to an equally wide variety of plant families has
yielded products with antibacterial properties. Such families include the Asteraceae,
Euphorbiaceae, Apocynaceae, Fabaceae, Leguminoceae, and Rutaceae [29].
Although no major antimicrobial drug has been developed from higher plants,
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 485
central role in complications of diabetes [45, 46]. Antioxidants play a major role in
protection against molecular oxidative damage [47]. In integration to oxidative
stress, insulin action is also impaired in diabetes, which leads to increased hepatic
glucose production. Due to the side effects of the existing synthetic drugs, plant-
derived food drugs are in great demand in industrially developed countries as alter-
native approach to treat diabetes. The WHO expert committee has recommended
that plants possessing hypoglycaemic activity may provide a utilizable source of
new oral antidiabetic drug for the development of pharmaceutical entities or may act
as simple dietary adjuncts to the existing therapies [48].
Until today, there is no treatment that can completely cure diabetes mellitus. In a
study by Maria et al. [49], it was found that insulin-dependent diabetes mellitus
(IDDM) or type I, otherwise, depends on insulin therapy to prevent the pathology
that would arise from insulin deficiency due to a dysfunction of the β-cells. However,
exogenous therapy which is the main treatment does not permit glycemic control as
precise as that provided by natural secretion from functional islet β-cells, and acute
decompositions and long-term complications are always present [50]. It is therefore
appropriate to look at possible alternative therapeutic strategies for IDDM. Type II
diabetes, otherwise known as adult-onset diabetes, results from a combination of
insulin resistance and inadequate secretion of insulin [51]. Management of type II
diabetes is rarely straightforward. It requires rigorous control of blood glucose and
special attention to a syndrome of associated vascular risk factors including hyper-
tension, dyslipidemia, and abdominal obesity. The pharmacological agents cur-
rently used for the 8 treatment of type II diabetes include sulfonylureas, biguanide,
thiazolidinedione, and α-glucosidase inhibitor. These agents, however, have
restricted use due to several undesirable side effects including haematological
effects, coma, and disturbances of the liver and kidney. In addition, they are not suit-
able for use during pregnancy [52]. Furthermore, the synthetic hypoglycemic agents
also fail to significantly alter the course of diabetic complications [53].
Therefore, the search for more effective and safer antidiabetic agents has contin-
ued to be an area of active research [54]. Moreover, studying the antidiabetic prop-
erty of herbal remedies will increase the chance of finding new antidiabetic drugs
and possibly finding a cheaper treatment for diabetes mellitus. Historical accounts
of diabetes mellitus first appeared in medical texts of several ancient cultures over
2000 years ago, and since ancient times, diabetes has been treated orally with sev-
eral medicinal plants. Ethnobotanical information indicates that more than 800
plants belonging to 28 families are used as traditional remedies for the treatment of
diabetes [55].
However, only a few of these plants have undergone comprehensive scientific
investigation, while 81% of these plants are hitherto unreported as antidiabetic
agents. Traditional medicines may be of considerable benefit especially during the
early stages of the illness, but most of the researchers worry about the side effect of
these products. Normally, herbs have ingredients with therapeutic activity, but their
preparation must be standardized to yield consistent products, which therefore can
be given in doses that are maximally safe and effective. In the absence of such stan-
dardization, the use of herbs in diabetes mellitus must be approached cautiously. So,
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 487
WHO has recommended that the research focus should now be directed to this area.
Sawarkar et al. [56] reported that numerous plants synthesize substances that are
useful in the maintenance of health in humans and animals. With a view to increas-
ing the wide range of medicinal uses, now the present day entails new drugs with
more potent and desired activity with lesser or no side effects against particular
disease [57].
The genus Ficus (Moraceae) constitutes one of the largest genera of angiosperms
includes with more than 800 species and 2000 varieties of Ficus genus, occur in the
most tropical and subtropical forests and this plant is commonly known as fig [58].
Sirisha et al. [59] reported that all Ficus species possess latex-like material within
their vasculatures that provide protection and self-healing from physical assaults.
Various studies indicated that Ficus species are widely used in the management of
various types of diseases like respiratory diseases, sexual disorders, central nervous
system diseases (CNS), cardiovascular disorders (CVS), gastric problems, skin
infections and diabetics, etc. [59].
Fig fruits hold the highest levels of polyphenols, flavonoids, and anthocyanins
and exhibited the highest antioxidant capacity, which can be free of side effects than
those of synthetic antioxidants [60]. Shukla et al. [61] revealed the significant anti-
oxidant effect of F. bengalensis. In addition, Duduku et al. [62] reported that
F. microcarpa bark contains highest free radical scavenging activity. Further, epide-
miological studies have shown that many of these antioxidant compounds possess
anti-inflammatory, analgesic, antimutagenic, anticarcinogenic, antibacterial, and
antiviral activities to a greater extent [63].
A large number of plants/plant extracts/decoctions or pastes are equally used by
tribal and folklore traditions in India for treatment of cuts, wounds, and burns [37].
Besides, in Ayurvedic medicine, F. racemosa L. is used as a wound-healing agent
[64], whereas the aqueous extract of F. deltoidea was reported to have wound-
healing activity [65].
Medicinal plants play an important role in cure of diabetes mellitus all over the
world. A variety of ingredients present in medicinal plants are thought to act on a
variety of targets by various modes and mechanisms. They have potential to impart
therapeutic effect in complicated disorders like diabetes and its complications [66].
According to Ayurvedic system of medicine, F. bengalensis, F. carica, F. glomerata
[67, 68], F. exasperate Vahl, and F. arnottiana Miq [69–71]. are well known in the
treatment of diabetes. F. carica [60], F. bengalensis extract of bark [72], and F. glom-
erata were reported to have hypoglycemic effect.
Moraceae, often called the mulberry family, is a family of flowering plants com-
prising of about 40 genera and over 1000 species of which over 800 species are
members of the genus Ficus (http://science.jrank.org/pages/4494/Mulberry-
FamilyMoraceae.html). They occur primarily in tropical and semitropical regions
488 S. P. Sasidharan et al.
and include a wide variety of herbs, shrubs, and trees, characterized by a milky sap
[73–75]. Moraceae have been divided into five tribes: Artocarpeae, Moreae,
Dorstenieae, Ficeae, and Castilleae [76]. Ficeae has only one genus, Ficus, with
approximately about 750 species largely distributed in 58 the tropics and subtrop-
ics [77]. They can be either monoecious with bisexual inflorescences or dioecious
[78]. Taxonomically, Ficeae is divided into two main groups [79]; one group, com-
prising the subgenera Urostigma and Pharmacosycea, consists of approximately
370 species, all of which are monoecious.
4 Genus Ficus
The genus Ficus L. (Moraceae) was first published in “Systema Naturae by Carolus
Linnaeus in 1735”. Ficus is one of the largest genus among angiosperms. Among
the genera of seed plants it ranked as the twenty-first [80]. The use of foods and
medicinal plants to improve health is nearly as old as humanity. Among such, none
may be older than the fig, which recent investigations have indicated has been culti-
vated for over 11,000 years, possibly predating cereal grains [81]. However, this
finding has recently been challenged [82]. A number of Ficus species are used as
food and for medicinal properties in Ayurvedic and Traditional Chinese Medicine
(TCM), especially among people where these species grow. These uses, however,
originated and are most widely found in the Middle East. It is reasonable to consider
that a survey of ethnomedical uses of a plant may provide useful clues for drug
discovery.
Ayurvedic medicine utilizes at least three different Ficus species [83, 84]. Figs are
used as external treatments for eczema, leprosy, rheumatism, sores, ulcers, and
pains. Like TCM, Ayurvedic medicine recommends gargling with fig decoctions for
sore throats and the ingestion of figs as a treatment for 59 diarrhea. Other Ayurvedic
uses include treatment of dysentery, gonorrhoea, and menorrhagia and as an aphro-
disiac [83]. In TCM, the fig is only rarely used, partly because this fruit is found
only in the most Southern regions of China, but also because the fruits are regarded
more as a food rather than a medicine and served after being salted like pickles,
cooked in water, or sun-dried. Medicinally, figs are recommended both for improv-
ing the appetite and as treatment for diarrhea. For hemorrhoids, a decoction of the
leaves is applied locally, while simultaneously cooked figs are also ingested orally.
Ficus religiosa (Peepal), F. benghalensis (Bar), F. benjamina (Sami), F. race-
mosa (Dumri), etc., possess high religious value for both Hindus and Buddhists [85]
and are deemed sacred. Ficus religiosa is not uprooted; it grows on shrines and
buildings, because it represents the Hindu god lord Vishnu, the god of sustenance. It
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 489
is widely worshipped as Bodhi tree under which lord Buddha attained enlighten-
ment [86].
Ficus benjamina is locally known as “Beringin, Waringin and Jejawi” [87]. It is
traditionally used as a stomachic, hypotensive, and anti-dysentry agent [88].
Previous studies on Ficus species revealed the presence of several compounds such
as alkaloids [89], triterpenes [90], ascorbic acid [91], and flavonoids [92].
Ficus exasperata is commonly known as sand paper tree and is spread in West
Africa in all kinds of vegetation and particularly in secondary forest regrowth. The
leaf extract from F. exasperata reported to have diverse uses such as treating hyper-
tensive patients [93], hemostative ophthalmia, coughs, and hemorrhoid [94]. In
Nigeria, young leaves of F. exasperata are prescribed as common anti-ulcer remedy.
Various pharmacological actions such as antidiabetic, lipid lowering, and antifungal
activities have been reported in F. exasperata [69].
F. bengalensis is the world’s largest tree in terms, its spread with some old trees
covering over an acre of ground. The tree’s name “banyan” refers to the merchants
who set up shop under the spreading trees. It is commonly found throughout India
and grows at deciduous and semi evergreen forests. F. bengalensis, known as Vata
in Sanskrit, is one of the reputed panchavalkala drugs of Ayurveda [95]. Different
parts of the tree have been found to possess medicinal properties; leaves are good
for ulcers, aerial roots are useful in gonorrhea, seeds and fruits are cooling and
tonic. The roots of F. bengalensis are given for obstinate vomiting and infusion of
its bark is considered as a tonic and astringent and is also used in diarrhea, dysen-
tery, and diabetes. Ayurvedic practitioners in India are using the milky juice (latex)
of F. bengalensis stem bark for the treatment of rheumatism and other inflammatory
diseases.
Ficus religiosa is commonly known as peepal in India. The plant is used to treat
gout, stomatitis, leucorrhea, ulcers, inflammation, and glandular swelling of the
neck [96]. F. religiosa has been reported for its wound healing [97], antibacterial
[98], and acetylcholinesterase inhibitory activities [99]. F. religiosa has been used
in the traditional system of Ayurveda to treat diabetes [100]. The leaves of F. reli-
giosa have been studied for antihyperglycemic activity [101].
F. glomerata Roxb. Syn. F. racemosa L. is commonly known as Gular in Hindi
and Cluster fig in English. It is medium sized to large evergreen or occasionally
deciduous tree and found all over India and Southeast Asia. Its fruits are mixed with
rice for making bread and used in several dishes. Traditionally, the bark, fruits, and
latex are used to treat anemia and gastrointestinal disorders like constipation and
dysentery [102].
F. racemosa L. (Indian fig) bark and fruits are well known to be useful in diabetes
[103] and found that Fistein and Resveratol were as effective as vitamin E in their
inhibitory action of lipid peroxidation. F. racemosa showed significant anticancer
[104] and antihelminthic activities [105]. The bark is antiseptic, antipyretic, and
vermicidal and a decoction of the bark is used in treating various skin diseases and
ulcers. It is used as a plaster in inflammatory swellings and boils. It is also effective
in the treatment of piles, dysentery, asthma, gonorrhea, hemoptysis, and urinary
diseases.
490 S. P. Sasidharan et al.
Several authors have isolated and identified various classes of compounds from the
genus Ficus. They include flavonoids, alkaloids, phenolic acids, steroids, saponins,
tannins, terpenoids, and coumarins (Fig. 1).
4.2.1 Steroids
β-sitosterol was isolated from the leaves and roots of F. carica and F. septica,
respectively. Stigmasterol has been reported in the leaves and roots of F. hirta and
F. septica [108]. 24-methylenecycloartenol, ψ-taraxasterol ester, lupeol, and baure-
nol have been isolated from the leaves of F. carica [109], whereas sitosterol-3-O-β-
D-glucoside has been reported from the leaves of F. septica [110].
β-sitosterol-α-D-glucoside has been isolated from the stem bark of F. benghalen-
sis [60].
4.2.2 Alkaloids
4.2.3 Coumarins
Another class of constituents commonly found in this genus are coumarins particu-
larly, of the furanocoumarin type. They have been isolated from the stem, leaves,
and roots of some Ficus species and are reported to be responsible for the contact
dermatitis associated with Ficus species [114]. The coumarins psoralen, bergapten,
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 493
4.2.4 Flavonoids
Genistin and kaempferitrin have been isolated from the leaves of F. septica [110].
Also, the leaves of F. ruficaulis and F. carica are reported to contain rutin and iso-
quercitrin [116]. Sheu et al. [118] reported the isolation of carpachromene, isogla-
branin, and norartocarpetin from the stem bark of F. formosana. Luteolin has also
been isolated from the aerial parts of F. pumilla [119]. The flavonoid apigenin has
been reported in the stem bark and roots of F. hirta and F. formosana. Also, the root
of F. hirta is reported to contain hesperidin, 5-hydroxy-41, 6, 7, 8- tetramethoxy
flavone, and 41, 5, 6, 7, 8-pentamethoxyflavone [108, 118]. Several other flavo-
noids, including alpinumisoflavone, cajanin, derrone, 5, 7-dihydroxy-4-methoxy-
31-(2,3-dihydroxy-3-methylbutyl) isoflavone, erycibenin A, erycibenin C, genistein,
31-(3-methylbut-2-enyl) biochanin A, and 5,7, 21- trihydroxy-41-methoxyisoflavone,
have been isolated from the stem bark of F. nymphaeifolia [120].
494 S. P. Sasidharan et al.
4.2.5 Triterpenoids
Squalene has been isolated from the leaves of F. septica [110]. Li et al. [108], iso-
lated 3β-acetoxy-α-amyrin, 3β-acetoxy-β-amyrin, and 3β-hydroxystigmast-5-en-7-
one from the roots of F. hirta. A number of triterpenoids with C-28 carboxylic acid
functional group have been isolated from the aerial roots of F. microcarpa. They
include betulonic acid, oleanonic acid, 3-oxofriedalan-28-oic acid, ursolic acid, and
ursonic acid [106]. Lupenol, lupenol acetate, 20-taraxastene-3β, 22α-diol, 29,
30-dinor-3β-acetoxy-18, 19-secolupane, 3, 22-dioxo-20- taraxastene, and
3β-hydroxy-20-oxo-29(20 → 19) abeolupane have also been isolated from the aerial
roots of F. microcarpa. Others include 3-acetoxy-12, 19-dioxo- 13(18)- oleanene,
3-acetoxy-1,11-epidioxy-12-ursene, 3-acetoxy-12,13-epoxy-11- hydroperoxyur-
sane, 3-acetoxy-11,12-epoxy-16-oxo-14-taraxerene, 3-acetoxy-11, 12- epoxy-14-
taraxerene, 3-acetoxy-20, 21-epoxytaraxastane, 3-acetoxy-21,
22- epoxytaraxastan-20-ol, 3-acetoxy20, 21-epoxytaraxastan-22-ol, and
3-acetoxy-18- hydroperoxy-12-oleanen-11-one [106].
4.2.6 Miscellaneous
Other compounds which have been isolated from this genus include bergenin and
racemosic acid from the stem bark of F. racemosa [108], cyanidin 3-O-glucoside
and cyanidin- 3-O-rhamnoglucoside from the fruits of F. carica [121], ficuformo-
diol A and ficuformodiol B from the stem bark of F. formosana [118].
4.3.1 Analgesic Activity
Analgesic activity of the leaf extract of Ficus glomerata Roxb. and stem bark of
F. bengalensis Linn has been confirmed, respectively, by Sehgal [122] and Patil and
Patil [71]. The ethanol extracts of F. racemosa bark and leaves were evaluated for
analgesic activity by analgesiometer at 100, 300, and 500 mg/kg and were found to
possess dose-dependent analgesic activity [123].
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 495
Anti-
inflammatory
Anti-microbial Anti-diabetic
Ficus Wound
Anti-cancer
species healing
Immunomodu
Anti-ulcer
latory
Analgesic
4.3.2 Anticancer Activity
4.3.3 Antiulcer Activity
The healing activity of whole plant extract of F. deltoidea Jack. was studied in gas-
tric ulcer induced by ethanol in rats; the extract promoted ulcer protection as ascer-
tained by the comparative significant decreases in ulcer areas and inhibition of
496 S. P. Sasidharan et al.
4.3.4 Antioxidant Activity
Cell membranes are especially vulnerable to the aggression of free radicals. When
the nucleus is damaged, the cell loses its ability to replicate itself. The impaired cell
replication results in the weakened immune system, skin aging, and many age-
related disorders. Various antioxidants deactivate the free radicals and prevent oxi-
dation on a cellular level. Some commonly used plants as antiaging agents include
F. bengalensis [72, 84].
The water extract (WE) and crude hot-water soluble polysaccharide (PS) from
F. carica fruit were investigated for scavenging abilities on DPPH, superoxide and
hydroxyl radicals, and reducing power. The immune activities of PS were evaluated
using the carbon clearance test and serum hemolysin analysis in mice. Both WE and
PS have scavenging activities on DPPH with the EC50 0.72 and 0.61 mg/ml, respec-
tively. The PS showed higher scavenging activity than WE on superoxide radical
(EC50, 0.95 mg/ml) and hydroxyl anion radical (scavenging rate 43.4% at 4 mg/ml).
The PS (500 mg/kg) also has a significant increase in the clearance rate of carbon
particles and serum hemolysin level of normal mice. This indicates the scavenging
activity and immune responses of the extract [60].
The antioxidant effect of species of Ficus may be attributed to the polyphenolic
compounds they possess. The antioxidant effect of aqueous extract of the bark of
F. bengalensis has been evaluated in hypercholesterolemia rabbits by Shukla et al.
[61] and confirmed its significant antioxidant effect. The potential health-promoting
constituents of fig fruits were studied with six commercial fig varieties differing in
color (black, red, yellow, and green) for total polyphenols, total flavonoids, antioxi-
dant capacity, and profile of anthocyanins. In the dark-colored mission and the red
Brown-Turkey varieties, the anthocyanin fraction contributed 36 and 28% of the
total antioxidant capacity, C3R (cyanidin- 3-O-rutinoside) contributed 92% of the
total antioxidant capacity of the anthocyanin fraction. Fruits of the mission variety
contained the highest levels of polyphenols, flavonoids, and anthocyanins and
exhibited the highest antioxidant capacity [60].
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 497
The ethanolic extract of leaves of Ficus religiosa was evaluated for antioxidant
(DPPH), wound healing (incision, excision, histopathological, and dead space
wound model), and anti-inflammatory (Carrageenan-induced paw edema) activity.
The tested extract of different dilutions in range 200 μg/ml to 1000 μg/ml shows
antioxidant activity in range of 6.34% to 13.35%. Significant increases in wound
closure rate, skin breaking strength, and granuloma breaking strength were observed.
The extract shows prominent anti-inflammatory activity as compared to that of stan-
dard (Ibuprofen gel) [128].
4.3.5 Antidiabetic Activity
The aqueous extract of Ficus religiosa Linn. was assessed at doses of 100 and
200 mg/kg orally and it decreased the fasting blood glucose in streptozotocin-
induced type 2 diabetic rats. The drug had enzyme induction effect with respect to
catalase (CAT) and glutathione peroxidase (GSHPx) activity, however, decreased
the exaggerated activity of superoxide dismutase (SOD) in type 2 diabetic rats.
F. religiosa modulated the enzymes of significant antioxidant defense system to
combat oxidative stress. Drug at higher dose (200 mg/kg) had more pronounced
effect. F. religiosa, a rasayana group of plant drug having antidiabetic activity along
with antioxidant potential, was beneficial in treatment of type-2 diabetes [129].
According to Ayurvedic system of medicine, F. bengalensis is well known in the
treatment of diabetes [67]. This attracted the attention of many earlier workers who
studied the hypoglycemic effect of extracts from its bark and tried to isolate active
compounds. Bark of this plant has antidiabetic properties. The hypoglycemic effect
of extract of bark was demonstrated in alloxan-induced diabetic rabbits, rats, and in
humans. Potent hypoglycemic water-insoluble principle was isolated from the bark
and a water-soluble hypoglycemic principle was also isolated from the bark which
was effective at a low dose of 10 mg/kg, bw/day [72]. Both the banyan bark prin-
ciples were effective in mild as well as severe alloxan-induced diabetes in rabbits
and improved lipid profile [130].
The aqueous leaf extract of F. carica induced a significant hypoglycaemic effect
in oral or intraperitoneal administration in streptozotocin - diabetic rats. Weight loss
was prevented in treated diabetic rats and the survival index was significantly altered
by plasma insulin levels [60]. Singh et al. [68] reported that F. bengalensis, F. car-
ica, and F. glomerata are effective in the treatment of diabetes. The hypoglycemic
activity of ethanol extracts of leaves of F. glomerata has significant antihyperglyce-
mic effect in experimental albino rat model of diabetes mellitus [71]. Hypolipidemic
effect of the water extract of the bark of F. bengalensis was investigated in alloxan-
induced diabetes mellitus in rabbits; showing a good glycemic control also corrects
the abnormalities in serum lipid profile associated with diabetes mellitus in view of
the ability of the extract to improve carbohydrate and lipid metabolism [61].
The fruits of F. glomerata, locally known as Gular, have been used since ancient
times in the ethnomedicine including as a remedy of diabetes mellitus [102]. The
aqueous extract of F. bengalensis at a dose of 500 mg/kg/day exhibits significant
498 S. P. Sasidharan et al.
4.3.6 Skin Diseases
As one of the oldest known human foods, figs as a fruit have a very high safety
profile. However, the toxicological evaluation of other fig products is in an early
stage. Skin contact with latex may provoke allergic reactions like dermatitis, asthma,
and anaphylaxis, while orally administered latex may induce hallucinosis [139].
Effects other than discussed above may be therapeutic or toxic, depending on the
clinical context [140].
4.3.7 Hepatoprotective Activity
The ethanol extract of F. carica, at doses of 100, 200, and 300 mg/kg, showed sig-
nificant dose-dependent reduction in normal body temperature and yeast-provoked
elevated temperature. The effect extended up to 5 h after drug administration when
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 499
4.3.8 Anthelmintic Activity
The latex of F. glabrata has been evaluated clinically and shown to be a potent and
well-tolerated anthelmintic agent [146], 1986). The methanolic, chloroform, and
petroleum ether extracts of the roots of F. bengalensis have potent anthelmintic
activity when compared with conventionally used drug and are equipotent to stan-
dard anthelmintic drug [147]. The aqueous extract of F. racemosa possesses wormi-
cidal activity and thus may be used as an anthelmintic [148].
4.3.9 Anti-inflammatory Activity
The anti-inflammatory effects of ethanolic and petroleum ether extracts of the bark
of F. bengalensis were evaluated in carrageenan-induced hind paw edema in rats
and the paw volume was measured plethysmometrically at 0 to 3 h after injection.
The results indicated that the ethanolic extract of F. bengalensis exhibited more
significant activity than petroleum ether in the treatment of inflammation compared
with the standard drug Indomethacin [72]. The methanolic extract of the stem bark
of Ficus religiosa L. was screened for its anti-inflammatory activity in Wistar albino
rats. A significant inhibition of carrageenan-induced rat paw edema, comparable to
500 S. P. Sasidharan et al.
that produced by indomethacin, was obtained with all the three doses of the extract.
The anti-inflammatory activity of F. racemosa extract was evaluated on carrageenan,
serotonin, histamine, and dextran-induced rat hind paw edema models where the
extract (400 mg/kg) exhibited anti-inflammatory effect of 30.4, 32.2, 33.9, and
32.0%, respectively. In a chronic test, the extract (400 mg/kg) showed 41.5% reduc-
tion in granuloma weight, which was comparable to that of phenylbutazone [144].
The acetone extract of Ficus amplissima leaves was studied for antioxidant activ-
ity, anti-inflammatory by using carrageenan and histamine-induced rat paw edema
models and wound healing activity. In this study, the acetone extract of Ficus
amplissima leaves exhibited better anti-inflammatory, wound healing, and in vivo
antioxidant activity probably due to phenols constituents [149]. In addition, the
effect of methanolic extract of the bark of Ficus amplissima (FAB) in streptozotocin-
induced diabetic rats and found that 50 mg/kg and 100 mg/kg than 150 mg/kg, three
doses, caused significant reduction in blood glucose levels. Also showed significant
increase in serum insulin and body weight, glycogen content in liver, skeletal mus-
cle, total protein contents, and significant reduction in the levels of serum triglycer-
ide and total cholesterol [150].
4.3.10 Immunomodulatory Activity
Gabhe [151] evaluated the immunomodulatory activity of the aerial roots of F. ben-
galensis. The successive methanol and water extracts exhibited a significant increase
in the percentage of phagocytosis versus the control. In the in vivo studies, the suc-
cessive methanol extract was found to exhibit a dose-related increase in the hyper-
sensitivity reaction to the SRBC antigen. It also resulted in a significant increase in
the antibody titter value to SRBC. The immunomodulatory effect of alcoholic
extract of the bark of F. religiosa in mice was investigated. Administration of extract
remarkably ameliorated both cellular and tic rats, while there was humoral antibody
response [152]. In Ayurvedic medicine, F. racemosa L. is used as a wound healing
agent [64]. The aqueous extract of the whole plant of F. deltoidea was investigated
by Abdulla et al. [65] to evaluate the rate of wound healing enclosure and the histol-
ogy of healed wounds in rats and results strongly document the beneficial and sig-
nificant effects to accelerate the rate of wound healing enclosure in the experimentally
induced wounds in rats. The wound-healing activity (incision and excision model)
of Ficus religiosa leaf extract prepared as ointment form (5 and 10%) was investi-
gated. Povidone iodine 5% was used as standard drug. The healing of wound was
assessed by the rate of wound contraction, decreased in the period of epithelializa-
tion, high skin breaking strength was observed in animals. Ten percent ointment
shows better wound healing activity than 5% concentration [57].
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 501
Fig trees of several different species show multiple cancer preventive, cancer thera-
peutic, and anti-inflammatory activities from their bark, roots, leaves, fruits, and
latex. Pharmacological and chemical studies have demonstrated antineoplastic or
anti-inflammatory activity of both the crude extract and pure compounds, of par-
ticular promise, due to their potent cytotoxic activity against a number of cancer cell
lines, which are the phenanthroindolizidine alkaloids and the triterpenoids with
C-18 carboxylic acid functional groups. In fact, these alkaloids, which have also
been found in a small number of other plant genera, are currently under active inves-
tigation as potential therapeutic leads [159]. In addition to these cytotoxic com-
pounds, several flavonoids, including anthocyanins, as well as other phenolic
compounds, demonstrated antioxidant and anti-inflammatory activities [108, 161].
502 S. P. Sasidharan et al.
Furthermore, lectins in the seeds may function as immune modulators [161]. The
sterols found in figs may also help bolster immunity, as well as inhibiting inflamma-
tion and invasion while promoting apoptosis and differentiation [162]. Coumarins,
in many cases, are selectively cytotoxic to cancer cells and also have antioxidant
activity and may interfere with formation of the lipoxygenase product 5-HETE to
suppress inflammation [163]. Some correlation between the ethnomedical employ-
ment and the pharmacological activities has been duly observed and noted in the
present review. As only a small fraction of the known Ficus spp. and parts has been
evaluated for anticancer and anti-inflammatory activities, figs may constitute a
rather large untapped source for new chemical entities with anticancer actions. In
last few years, there has been an exponential growth in the field of herbal medicines
to cure various diseases including diabetes.
Ficus species have been identified as a potential and significant medicinal plant used
to treat a variety of diseases, according to a comprehensive literature assessment.
The world is abundant in therapeutic herbs, which is quite pleasurable. The scien-
tific work of the Ficus genus was reviewed in this article. The traditional uses, phy-
tochemical components, and medicinal action of these plants were discussed. The
goal of this research is to compile a list of articles on Ficus species. Secondary
metabolites such as triterpenoid, steroid, saponin, flavonoid, phenolic compound,
and alkaloid were discovered in various Ficus species, according to a critical study
of the literature. Quercetin, quercetin 3-O—L-arabinopyranoside, epilupeol acetate,
oleanolic acid, friedelin, elastiquinone, pinocembrin-7-O—D-glucoside, and ficu-
soside B were all isolated as pure compounds.
These opportunities derive primarily from the extremely benign safety profile of
fig fruit products, pleasant taste, and its antioxidant constituents. Because the anti-
oxidant action is also a means of lowering chronic anti-inflammatory action and
insulin resistance, fig fruits hold potential in functional food approaches aimed at
normalizing metabolic syndrome and boosting wellness beyond the widely accepted
role of figs in the diet for improving bowel performance and as a source of naturally
sweet, readily available, quick energy. Ficus species are rich source of polyphenolic
compounds, flavonoids, which are responsible for strong antioxidant properties that
help in prevention and therapy of various oxidative stress-related diseases such as
pain reliever, cancer, anti-ulcerogenic, aging, diabetes, fever, antherogenesis, hel-
minths infections, inflammation, immune system, diarrhea, allergy, and stress [160].
Traditional uses, phytochemical components, and medicinal activities of Ficus
plants were summarized. The majority of the species were utilized as traditional
medicine in Asian nations such as Indonesia, Papua New Guinea, Vietnam, Pakistan,
Thailand, and Vanuatu, according to the literature review. Some Ficus species
require additional research into their pharmacological properties, based on pro-
cesses and chemical content.
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 503
References
1. Omonkhelin JO, Eric KIO, Osahon O (2007) Antifungal and antibacterial activities of the
ethanolic and aqueous extract of Kigelia africana (Bignoniaceae) stem bark. Afr J Biotechnol
6(14):1677–1680
2. De Pasquale A (1984) Pharmacognosy: the oldest modern science. J Ethnopharmacol 11:1–16
3. Rates SMK (2001) Plants as source of drugs. Toxicon 39:603–613
4. Ngo LT, Okogun JI, William R (2013) Folk 21st century natural product research and drug
development and traditional medicines. Nat Prod Rep 30:584
5. Chevalier A (1995) Encyclopedia of medicinal plants. Amazon Press, pp 3–8
6. Houghton PL (1995) The role of plants in traditional medicine and current therapy. J Altern
Complement Med 1:131–143
7. Shu Y-Z (1998) Recent natural products based drug development: a pharmaceutical industry
perspective. J Nat Prod 61:1053–1071
8. Hamburger M, Hostettmann K (1991) Bioactivity in plants: the link between phytochemistry
and medicine. Phytochemistry 30(12):3864–3874
9. Verpoorte R (2000) Pharmacognosy in the new millenium: leadfinding and biotechnology. J
Pharm Pharmacol 52:253–262
10. Cadenas E, Packer L (2002) Handbook of antioxidants. Marcel Dekker, New York
11. McCord JM (2000) The evolution of free radicals and oxidative stress. Am J Med 108:652
12. Freidovich I (1999) Fundamental aspects of reactive oxygen species, or what’s the matter
with oxygen? N.Y. academic. Science 893:13
13. Yun-Zhong F, Sheng Y, Guoyao W (2002) Free radicals, antioxidants, and nutrition. Nutrition
18:872–879
14. Emery P (2006) Treatment of rheumatoid arthritis. Br Med J 332:152–155
15. Willerson JT, Ridker PM (2004) Inflammation as a cardiovascular risk factor. Circulation
109, II:2–10
16. Rang HP, Dale MM (1991) The endocrine system pharmacology, Second edn. Longman
Group Ltd, pp 504–508
17. McGettigan MP, Henry D (2000) Current problems with non-specific COX inhibitors. Curr
Pharm Des 6:1693–1724
18. Derle DV, Gujar KN, Sagar BSH (2006) Adverse effect associated with the use of nonsteroi-
dal anti-inflammatory drugs: an overview. Indian J Pharm Sci 68:409–414
19. Lin CR, Amaya F, Barrett L, Wang H, Takada J, Samad T, Woolf CJ (2006) Prostaglandin E2
receptor EP4 contributes to inflammatory pain hypersensitivity. J Pharmacol Experimental
Therap 319:1096–1103
20. Dharmasiri JR, Jayalcody AC, Galhena G, Liyanage SSP, Ratansooriya WD (2003)
Antiinflammatory and analgesic activities of mature fresh leaves of Vitex negundo. J
Ethanopharmacol 87:199–206
21. Berman B (2004) Willow bark. University of Maryland Medical Center, Baltimore. http://
www.umm.edu/altmed/Cons Herbs/WillowBarkch.html
22. Satoskar RR (1996) Evaluation of anti-inflammatory property of curcumin in patients with
post-operative inflammation. Int J Clin Pharmacol Ther Toxicol 24:651–654
23. Srivastava KC, Mustafa T (1992) Ginger (Zingiber officinale) in rheumatism and musculo-
skeletal disorders. Med Hypotheses 39:342–348
24. Boon H, Smith M (1997) Natural anti-inflammatory supplements: research status and clinical
applications. In: Meschino DJ (ed.) Health care professional training program in complemen-
tary medicine
25. Wu SJ, Tsai JY, Chang SP, Lin DL, Wang SS, Huang SN, Ng LT (2006) Supercritical carbon
dioxide extract exhibits enhanced antioxidant and anti-inflammatory activities of Physalis
peruviana. J Ethnopharmacol 108:407–413
504 S. P. Sasidharan et al.
26. Chi YS, Jong HG, Son KH, Chang HW, Kang SS, Kim HP (2001) Effects of naturally occur-
ring prenylated flavonoids on enzymes metabolising arachidonic acid: lipoxygenases and
cyclooxygenases. Biochem Pharmacol 62:1185–1191
27. Eleni P, Dimitra HL (2003) Review in quantitative structure activity relationships on lipoxy-
genase. Mini-Rev Med Chem 3:487–499
28. Robbers J, Speedie M, Tyler V (1996) Pharmacognosy and pharmacobiotechnology. Williams
and Wilkins, Baltimore, pp 1–14
29. Roy A, Saraf S (2006) Ethnomedicinal approach in biological and chemical investigation of
phytochemicals as antimicrobials. Pharm Rev 4(2)
30. Ellof JN (1990) It is possible to use herbarium specimens to screen for antibacterial compo-
nents in some plants. J Ethnopharmacol 67:355–360
31. Gurib-Fakim A (2006) Medicinal plants: tradition of yesterday and drugs of tomorrow. Rev
Article Mol Aspects Med 27(1):1–93
32. Senthilkumar GP, Arulselvan P, Sathishkumar D, Subramanian SP (2006) Antidiabetic
activity of fruits of Terminalia chebula on streptozotocin induced diabetic rats. J Health Sci
52:283–291
33. Meenakshi S, Raghavan G, Nath V, Ajay Kumar SR, Shanta M (2006) Antimicrobial,
wound healing and antioxidant activity of Plagiochasma appendiculatum Lehm. Et Lind. J
Ethnopharmacol 107:67–72
34. Enoch S, John LD (2005) Basic science of wound healing. Surgery 23:37–42
35. Sumitra M, Manikandana P, Suguna L (2005) Efficacy of Butea monosperma on dermal
wound healing in rats. Int J Biochem Cell Biol 37:566–573
36. Suresh RJ, Rao PR, Reddy MS (2002) Wound healing effects of Heliotropium indicum,
Plumbago zeylanicum and Acalypha indica in rats. J Ethnopharmacol 79:249–251
37. Kumar B, Vijayakumar M, Govindarajan R, Pushpangadan P (2007) Ethnopharmacological
approaches to wound healing – exploring medicinal plants of India. J Ethnopharmacol
114:103–113
38. Krishnan P (2006) The scientific study of herbal wound healing therapies: current state of
play. Curr Anaesth Crit Care 17:21–27
39. Mohamed AK, Bierhaus A, Schiekofer S, Tritschler H, Ziegler R, Nawroth PP (1999) The
role of oxidative stress and NF-κB activation in late diabetic complications. Biofactors
10:157–167
40. Halliwell B, Gutteridge JMC (1989) Free radicals, other reactive species and disease. In: Free
radicals in biology and medicine. Clarendon Press, Oxford, pp 617–623
41. Baynes JW (1991) Perspective in diabetes. Role of oxidative stress in development of com-
plication in diabetes. Diabetes 40:405–412
42. Ceriello A (2000) Oxidative stress and glycemic regulation. Metabolism 49:27–29
43. Fatima SS, Rajasekhar MD, Kumar KV, Kumar MTS, Babu KR, Rao CA (2010) Antidiabetic
and antihyperlipidemic activity of ethyl acetate:isopropanol (1:1) fraction of Vernonia anthel-
mintica seeds in streptozotocin induced diabetic rats. Food Chem Toxicol 48:495–501
44. WHO (World Health Organization) (2009) Prevalence data of diabetes world wide
45. Dewanjee S, Das AK, Sahu R, Gangopdhyay M (2009) Antidiabetic activity of Diospyros
peregrine fruit: effect on hyperglycemia, hyperlipidemia and augmented oxidative stress in
experimental type 2 diabetes. Food Chem Toxicol 47:2679–2685
46. FAO/WHO (1990) Energy and protein requirements. Report of joint FAO/WHO/UNU Expert
Consultation Technical Report. FAO/WHO and United Nations University, Geneva
47. Evans JL (2007) Antioxidants: do they have a role in the treatment of insulin resistance.
Indian J Med Res 125:355–372
48. WHO (1980) Expert committee on diabetes mellitus second report. Technical Report Series
646. World Health Organization, Geneva, p 61
49. Maria AB, Assimina GT, Drakoulis D, Alexandra F, Panayiotis HP (1999) Salivary altera-
tions in insulin-dependent diabetes mellitus. Int J Paediatr Dent 8(1):29–33
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 505
50. Larsen MO, Raun K, Ribel U, Gotfredsen CF, Brand CL, Wilken M, Carr RD, Rolin B (2003)
Insulin sensitivity is negatively correlated to total body mass in Göttingen minipigs. Diab
Metab 29:S98
51. Reaven G (1988) Role of insulin resistance in human disease. Diabetes 37:1595–1607
52. Pari L, Uma M (1999) Hypoglycaemic effect of Musa sapientum L. in alloxaninduced dia-
betic rats. J Ethnopharmacol 68:321–325
53. Rang HP, Dale MM, Ritter JM (1999) Anti-inflammatory and immunosuppressant drugs. In:
Pharmacology, 5th edn. Churchil Livingstone Edinburgh, London, p 248
54. Pari L, Venkateswaran S (2002) Hypoglycaemic activity of Scopariadulcis L. extract in
alloxan induced hyperglycaemic rats. Phytother Res 16(7):662–664
55. Eidi M, Eidi A, Zamanizadeh H (2005) Effect of Salvia officinalis L. leaves on serum glu-
cose and insulin in healthy and streptozotocin induced diabetic rats. J Ethnopharmacol
100:310–313
56. Sawarkar HA, Singh MK, Pandey AK, Biswas D (2011) In vitro anthelmintic activity of
Ficus bengalhensis, Ficus caria and Ficus religiosa: a comparative anthelmintic activity. Int J
Pharm Tech Res 3:152–153
57. Roy K, Shivkumar H, Sarkar S (2009) Wound healing potential of leaf extracts of Ficus reli-
giosa on Wistar albino strain rats. Int J Pharm Tech Res 1(3):506–508
58. Hamed MA (2011) Beneficial effect of Ficus religiosa Linn. On highfat- induced hypercho-
lesterolemia in rats. Food Chem 129:162–170
59. Sirisha N, Sreenivasulu M, Sangeeta K, Chetty CM (2010) Antioxidant properties of Ficus
species-A review. Int J PharmTech Res 4:2174–2182
60. Joseph B, Raj SJ (2011a) Pharmacognostic and phytochemical properties of Ficus carica
Linn –an overview. Int J PharmTech Res 3(1):8–12
61. Shukla R, Gupta S, Gambhir JK, Prabhu KM, Murthy PS (2004) Antioxidant effect of aqueous
extract of the bark of Ficus bengalensis in hypercholesterolaemic rabbits. J Ethnopharmacol
92:47–51
62. Duduku K, Rosalam S, Leenah NJ (2007) Recovery of phytochemical components from
various parts of Morinda citrifolia extracts by using membrane separator. J Appl Sci
7(15):2093–2098
63. Owen RW, Giacosa A, Hull WE, Haubner R, Spiegelhalder B, Bartsch H (2000) The anti-
oxidant/anticancer potential of phenolic compounds isolated from olive oil. Eur J Cancer
36:1235–1247
64. Biswas TK, Mukherjee B (2003) Plant medicines of Indian origin for wound healing activity:
a review. Int J Lower Extreme Wounds 2(1):25–39
65. Abdulla MA, Ahmed KA, Abu-Luhoom FM, Muhanid M (2010) Role of Ficus deltoidea
extract in the enhancement of wound healing in experimental rats. Biomed Res 21(3):241–245
66. Tiwari AK, Rao MJ (2002) Diabetes mellitus and multiple therapeutic approaches of phyto-
chemicals: present status and future prospects. Curr Sci 83:30–38
67. Rashid ABA (2008) The chemical constituents from the stems of Ficus deltoids. B.Sc. Final
Year Project Report. University Teknologi Mara, Selangor
68. Singh S, Gupta SK, Sabir G, Gupta MK, Seth PK (2009) A database for antidiabetic plants
with clinical/experimental trials. Bioinformation 4(6):263–268
69. Sonibare MO, Isiaka AO, Taruka MW, Williams NS, Soladoye M, Emmanuel O (2006)
Constituents of Ficus exasperata leaves. Nat Prod Commun:23–26
70. Mazumdar PM, Farswan M, Parcha V (2009) Hypoglycaemic effect of Ficis arnottiana Miq.
Bark extracts on streptozotocin induced diabetea in rats. Nat Prod Radiance 8(5):478–482
71. Sharma VK, Kumar S, Patel HJ, Hugar S (2010) Hypoglycemic activity of Ficus glomerata in
alloxan induced diabetic rats. Int J Pharmaceut Sci Rev Res 1(2):18–22
72. Patil VV, Patil VR (2010) Ficus Benghalensis Linn.-an overview. Int J Pharm Bio Sci 6:2
73. Everett TH (1968) Living trees of the world. Doubleday, New York. Retrieved at (http://sci-
ence.jrank.org/pages/4494/Mulberry-Family-Moraceae.ecologyeconomic- value.html)
506 S. P. Sasidharan et al.
74. Duncan WH, Duncan MB (1988) Trees of the southeastern United States. University
of Georgia Press. Retrieved at http://science.jrank.org/pages/4494/Mulberry-Family-
Moraceae.html
75. Godfrey RK (ed) (1988) Trees, shrubs, and Woody vines of northern Florida and adja-
cent Georgia and Alabama. University of Georgia Press. Retrieved at http://science.jrank.
org/pages/4493/Mulberry-Family-Moraceae-Ecologydistribution-economic-value.html on
29/09/09
76. Rohwer JG, Kubitzki K, Bittrich V (eds) (1993) The families and genera of vascular plants.
Springer Verlag, Berlin, Germany, pp 438–453
77. Shannon L, Datwyler and Weiblen, G.D. (2004) On the origin of the fig: phylogenetic rela-
tionships of moraceae from NDHF sequences. Am J Bot 91(5):767–777
78. Weiblen GD (2000) Phylogenetic relationships of functionally dioecious Ficus (Moraceae)
based on ribosomal DNA sequences and morphology. Am J Bot 87:1342–1357
79. Berg CC, Hijman MEE (1989) Moraceae. In: Polhill RM (ed) Flora of tropical East Africa.
Rotterdam, Netherlands, p 95
80. Lansky EP, Paavilainen HM, Pawlus AD, Newman RA (2008) Ficus spp. (fig): ethnobotany
and potential as anticancer and anti-inflammatory agents. J Ethnopharmacol 119:195–213
81. Kislev ME, Hartmann A, Bar-Yosef O (2006) Early domesticated fig in the Jordan Valley.
Science 312:1372–1374
82. Lev-Yadun S (2006) Defensive coloration in plants: a review of current ideas about anti-
herbivore coloration strategies. In: Teixeira da Silva JA (ed) Floriculture, ornamental and
plant biotechnology. Advances and topical issues, vol VI. Global Science Books Ltd, London,
UK, pp 292–299
83. Kapoor LD (1990) CRC handbook of Ayurvedic medicinal plants. CRC Press, BocaRaton
84. Khan I, Alam S, Akhter S, Shahin N, Ansari FZ (2007) Ageing and its herbal treatment.
Pharma Rev 12:131–134
85. Subedi A, Paudyal G (2001) Some notable orchid of Pokhara valley and their habitats.
Botanica Orientalis 2:172–174
86. Majupuria TC, Joshi DP (1989) Religious and useful plants of Nepal and India. M. Gupta,
Lashkar, India
87. Holttum RE (1969) Plant life in Malaya. London Longman Group Limited, p 87
88. Trivedi CP, Shinde S, Sharma RC (1969) Prelimnary phytochemical and pharmacological
studies on Ficus racemosa extract (gular). Indian J Med Res 57(6):1070–1074
89. Beat B, Clemens AJ, Wright D, Rali T, Orro S (1990) An antimicrobial alkaloid from Ficus
septic. Phytochemistry 29(10):3327–3330
90. Mohammad A, Sutradhar A, Ahmad M, Ranjit K (1991) Chemical constituents of Ficus
glomerata Roxb. J Bangaladesh Chem Soc 4(2):247–250
91. Ikhals AK, Rali T, Sticher O (1993) Alkaloids from Ficus pachyrhachis. Planta Med 59(3):286
92. Ilyas M, Ilyas N (1990) Flavonids from the leaves of Ficus capensis. Ghana J Chem
1(3):176–178
93. Buniyamin AA, Eric KIQ, Fabian CA (2007) Pharmacognosy and hypotensive evaluation
of Ficus exasperata Vahl.(moraceae) leave. Acta Poloniae Pharm Drug Res 64(6):543–546
94. Odunbaku OA, llusanya O.A., Akasoro K.S. (2008) Antimicrobial activity of ethanolic leaf
extract of Ficus exasperata on Escherchia coli and staphylococcus albus. Sci Res Essay
3(11):562–564
95. Shantha TR, Shetty JK, Indira A, Bikshapathi T (2006) Pharmacognostical studies on Vata
shrung (Ficus benghlensis Linn. Leaf primordium). Ind J Trad Knowledge 5(3):388–393
96. Kirtikar KR, Basu BD (1933) Indian medicinal plants, vol 1–4. Publisher L M Basu,
Allahabad
97. Naira N, Rohini R, Asdag S, Das A (2009) Wound healing activity of the hydro alcoholic
extract of Ficus religiosa leaves in rats. Int J Alternative Med 6:2
98. Aqil F, Ahamd I (2003) Broad spectrum antibacterial and antifungal properties of certain
traditionally used Indian medicinal plants. World J Microbiol Biotechnol 19:653–657
An Overview of Ethnobotany, Phytochemicals, and Pharmacological Properties… 507
99. Vinutha B, Prashanth D (2007) Screening of selected Indian medicinal plants for acetylcho-
lineesterase inhibitory activity. J Ethnopharmacol 109:359–363
100. Simmonds M, Howes M (2006) Plants used in the treatment of diabetes. In: Soumyanath A
(ed) Traditional medicines for modern time-antidiabetic plants, vol 6. CRC Press/Taylor and
Francis Group, pp 19–82
101. Deshmukh T, Yadav B, Badole Bodhankar S, Dhaneshwar S (2007) Antihyperglycemic activ-
ity of alcoholic extract of Ficus religiosa leaves in alloxian induced diabetic mice. J Herbal
Med Toxicol 1:80–86
102. Chopra RN, Nayar SL, Chopra IC (2002) Glossary of Indian medicinal plants. Council of
Scientific and Industrial Research, NISCAIR, New Delhi, p 199
103. Bhardwaj, A., Hundal, M.K., Pathania, V., Syal, N., Sohi, K.K., Khanduja, K.L., (2000).
Evaluation of antioxidant and antiradical activity of reveratrol, paper presented at the 19th
annual convention of Indian association for cancer research and symposium on cancer biol-
ogy, 21–23
104. Rubnov S, Kashman Y, Rabinowitz R, Schlesinger Mechoulam R (2001) Suppressors of can-
cer cell proliferation from fig (Ficus carica) resin; isolation and structure elucidation. J Nat
Prod 64:993–996
105. Hansson A, Veliz G, Naquira C, Amren M, Arroyo M, Arevalo G (1986) Preclinal and clini-
cal studies with latex from Ficus glabrata HBK, a traditional intestinal anthelinthic in the
Amazonian area. J EthnopharmacolJournal of Ethnopharmacology 2:105–138
106. Chiang YM, Chang JY, Kuo CC, Chang CY, Kuo YH (2005) Cytotoxic triterpenes from the
aerial roots of Ficus microcarpa. Phytochemistry 66:45–501
107. Baumgartner B, Erdelmeier CAJ, Wright AD, Rali T, Sticher O (1990) An antimicrobial
alkaloid from Ficus septica. Phytochemistry 29:3327–3330
108. Li C, Bu PB, Yue DK, Sun YF (2006) Chemical constituents from roots of Ficus hirta.
Zhongguo Zhong YaoZaZhi 31:131–133
109. Saeed MA, Sabir AW (2002) Irritant potential of triterpenoids from Ficus carica leaves.
Fitoterapia 73:417–420
110. Wu PL, Rao KV, Su CH, Kuoh CS, Wu TS (2002) Phenanthroindolizidine alkaloids and their
cytotoxicity from the leaves of Ficus septica. Heterocycles 57:2401–2408
111. Damu AG, Kuo PC, Shi LS, Li CY, Kuoh CS, Wu PL, Wu TS (2005) Phenanthroindolizidine
alkaloids from the stems of Ficus septica. J Nat Prod 68:1071–1075
112. Yang CW, Chen WL, Wu PL, Tseng HY, Lee SJ (2006) Anti- inflammatory mechanisms of
phenanthroindolizidine alkaloids. Mol Pharmacol 69:749–758
113. Venkatachalam SR, Mulchandani NB (1982) Isolation of phenanthroindolizidine alkaloids
and a novel biphenylhexahydroindolizine alkaloid from Ficus hispida. Naturwissenschaften
69:287–288
114. Zaynoun ST, Aftimos BG, Abi AL (1984) Ficus carica; isolation and quantification of the
photoactive components. Contact Dermatitis 11:21–25
115. Innocenti G, Bettero A, Caporale G (1982) Determination of the coumarinic constituents of
Ficus carica leaves by HPLC. Farmaco 37:475–485
116. Chang YF, Chi CW, Chern YT, Wang JJ (2005) Effects of 1,6-Bis[4-(4-amino3- hydroxyphe-
noxy)phenyl]diamantane (DPD), a reactive oxygen species and apoptosis inducing agent, on
human leukemia cells in vitro and in vivo. Toxicol Appl Pharmacol 202:1–12
117. Makhija IK, Sharma IP, Khamar D (2010) Phytochemistry and pharmacological properties of
Ficus religiosa: an overview. Ann Biol Res 1(4):171–180
118. Sheu YW, Chiang LC, Chen IS, Chen YC, Tsai IL (2005) Cytotoxic flavonoids and new
chromenes from Ficus formosana. Planta Med 71:1165–1177
119. Pistelli L, Chiellini EE, Morelli I (2000) Flavonoids from Ficus pumila. Biochem Syst Ecol
28(3):287–289
120. Darbour N, Bayet C, Rodin-Bercion S, Elkhomsi Z, Lurel F, Chaboud A, Guilet D (2007)
Isoflavones from Ficus nymphaeifolia. Nat Prod Res 21:461–464
508 S. P. Sasidharan et al.
145. Channabasavaraj KP, Badami S, Bhojraj S (2008) Hepatoprotective and antioxidant activity
of methanol extract of Ficus glomerata. J Nat Med 62:379–383
146. Morton AD, Mc Manus IC (1986) Attitudes to and knowledge about the acquired immune
deficiency syndrome: lack of a correlation. Br Med J 293:1212
147. Aswar M, Aswar U, Watkar B, Vyas M, Wagh A, Gujar KN (2008) Anthelmintic activity of
Ficus bengalensis. Int J Green Pharm 2(3):170–172
148. Chandrashekhar CH, Latha KP, Vagdevi HM, Vaidya VP (2008) Anthemintic activity of the
crude extracts of Ficus racemosa. Int J Green Pharm 2:100–103
149. Arunachalam K, Parimelazhagan T (2013) Anti-inflammatory, wound healing and in-
vivo antioxidant properties of the leaves of Ficus amplissima Smith. J Ethnopharmacol
145(1):139–145
150. Arunachalam K, Parimelazhagan T (2013) Antidiabetic activity of Ficus amplissima Smith.
Bark extract in streptozotocin induced diabetic rats. J Ethnopharmacol 147(2):302–310
151. Gabhe SY, Tatke PA, Khan TA (2006) Evaluation of the immunomodulatory activity of the
methanol extract of Ficus bengalensis roots in rats. Indian J Pharmacol 38(4):271–275
152. Mallurwar VR, Pathak AK (2008) Studies on immunomodulatory activity of Ficus religiosa.
Indian J Pharm Educ Res 42(4):341–343
153. Mukherjee PK, Saha K, Murugesan T, Mandal SC, Pal M, Saha BP (1998) Screening of
anti-diarrhoeal profile of some plant extracts of a specific region of West Bengal. India J
Ethnopharmacol 60:85–89
154. Taur DJ, Nirmal SA, Patil RY, Kharya MD (2007) Antis tress and ant allergic effects of Ficus
bengalensis bark in asthma. Nat Prod Res 21(14):66–70
155. Malhotra CL, Das PK, Dhalla NS (1960) Parasympatholytic activity of Ficus religiosa Linn.
Indian J Med Res 48:734–742
156. Vinson JA, Zubik L, Bose P, Samman N, Proch J (2005) Dried fruits: excellent in vitro and
in vivo antioxidants. J Am Coll Nutr 24(1):44–50
157. Serraclara A, Hawkins F, Peez C, Dominguez E, Campillo JE, Torres MD (1998)
Hypoglycemic action of an oral fig-leaf decoction in type-1 diabetic patients. Diabetes Res
Clin Pract 39:19–22
158. Bohlooli S, Mohebipoor A, Mohammadi S, Kounhavard M, Pashapoor S (2007) Comparative
study of fig tree efficacy in the treatment of common warts (verruca vulgarisvs. Cryotherapy).
Int J Dermatol 46:524–526
159. Gao W, Lam W, Kaczmarek C, Baker DC, Cheng YC (2004) Novel mode of action of tyloph-
orine analogs as antitumor compounds. Cancer Res 64:678–688
160. Priya D, Purnima D, Borthakur SK (2013) Pharmaceutical properties of Indian species of
Ficus Linn. Int J Pharm Life Sci 4(1):2314–2319
161. Rabinovich GA, Liu FT, Hirashima M, Anderson A (2007) An emerging role for galectins
in tuning the immune response: lessons from experimental models of inflammatory disease,
autoimmunity and cancer. Scand J Immunol 66:143–158
162. Bradford PG, Awad AB (2007) Phytosterols as anticancer compounds. Mol Nutri Food Res
51:161–170
163. Kulkarni MV, Kulkarni GM, Lin CH, Sun C (2006) Recent advances in coumarins and
1-azacoumarins as versatile biodynamic agents. Curr Med Chem 13:2795–2818
Phytochemistry and Pharmacology
of Catharanthus roseus (L.) G. Don
and Rauvolfia serpentina (L.) Benth. ex
Kurz
1 Introduction
Catharanthus roseus (L.) G.Don and Rauvolfia serpentina (L.) Benth. ex Kurz are
important and the most explored plant species belonging to the dogbane family
Apocynaceae. These species recorded as endemic to Madagaskar and originate
from India (Fig. 1). C. roseus is commonly known as pink periwinkle, rose peri-
winkle, Cape periwinkle, bright eyes, Madagascar periwinkle, graveyard plant, and
old maid, whereas Indian snakeroot, sarpagandha, Chandrika and devil pepper, or
serpentine wood are common names of R. serpentina [1–5]. In the Ayurvedic sys-
tem of medicine, different parts of C. roseus and R. serpentina are used in folklore
herbal medicine for the treatment of many types of cancer, diabetes, stomach disor-
ders, kidney, liver, snakebites, hypertension, and associated cardiovascular diseases
by the local people of tropical regions of Africa, Asia, America, and various Oceanic
islands [6–11]. C. roseus is an ornamental plant that has flowers of a different color
for most of the year. Both the plant species have been distributed throughout the
world and used in traditional medicine for ages in various parts of the world [11].
The extracts, fractions and compounds of C. roseus and R. serpentina plants are
extensively studied for their pharmacological activities such as antimicrobial, anti-
oxidant, antiprotozoal, antitrypanosomal, antipsychotic, cardioprotective, cholines-
terase inhibitory, and hepatoprotective due to the rich content of bioactive
S. Kumar
Department of Chemistry, Ma. Kanshiram Government Degree College, Ninowa (affiliated to
Chhatrapati Shahu Ji Maharaj University (CSJM) Kanpur), Farrukhabad, Uttar Pradesh, India
B. Singh (*)
Botanic Garden Division, CSIR-National Botanical Research Institute, Rana Pratap Marg,
Lucknow, Uttar Pradesh, India
Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
e-mail: [email protected]
Fig. 1 Pictures of Catharanthus roseus (a), Rauvolfia serpentina (b), and Rauvolfia serpentina
root (c)
2 Phytochemistry
The biosynthesis of indole alkaloids in MIAs is very complex, involving more than
50 metabolic steps to synthesize the most important alkaloids (Fig. 2). About 20 of
the 50 enzymes necessary for their biosynthesis have been identified and character-
ized by different techniques. Thus, there are still a number of important enzymes
that are essential to be characterized, which can only be done after the isolation and
Table 1 Different classes of Secondary metabolites with mass, molecular formula, their distribution in parts, and pharmacological properties of Catharanthus
roseus and Rauvolfia serpentina
Molecular
Serial no. Compound/subclass MS (Da) formula Distribution or source Species Activity
1. Sitsirikine 354 C21H26N2O3 Leaves, stem bark, and C. roseus, R. Immunosuppressive
twigs serpentina
2. 10-Methoxynormacusine 324 C20H24N2O2 Leaves, stem bark, root R. serpentina Immunosuppressive
B (Lochnerine) (10-O- bark
Methylsarpagine)
3. Tetraphyllicine 308 C20H24N2O Leaves, stem bark, and C. roseus, R. Myocardial excitation
(serpinine) root bark serpentina
4. Ajmaline 326 C20H26N2O2 Leaves, stem bark, and C. roseus, R. Antiarrhythmia, antifibrillation, cardiac and liver
root bark serpentina toxicity, immunosuppressive
5. Isoreserpiline 412 C23H28N2O5 Leaves, stem bark, and C. roseus, R. Antipsychotic
root serpentina
6. Reserpiline 412 C23H28N2O5 Leaves, stem bark, and C. roseus, R. Antipsychotic, antigastric secretion,
root serpentina antihypertension
Molecular
Serial no. Compound/subclass MS (Da) formula Distribution or source Species Activity
12. Isorauhimbinic acid 340 C20H24N2O3 Root C. roseus, R. Cytotoxic
serpentina
13. α-Yohimbine 354 C21H26N2O3 Leaves, stem bark, and C. roseus, R. Immunosuppressive, antibacterial, cytotoxic,
root serpentina pre- and postsynaptic α2-adrenoceptor inhibitor,
stimulant and aphrodisiac effect, antidiabetic
14. β-Yohimbine 354 C21H26N2O3 Leaves, stem bark, and C. roseus, R. Cytotoxic
root serpentina
15. 18-Hydroxyyohimbine 370 C21H26N2O4 Stem, stem bark, and C. roseus, R. Antipsychotic, antihypertension, antidepression,
root bark serpentina anticancer, carcinogenesis
16. Reserpine 608 C33H40N2O9 Leaves, stem bark, and C. roseus, R. Immunosuppressive
root serpentina
17. Rescinnamine 634 C35H42N2O9 Leaves and root bark R. serpentina Antihypertension
18. Akuammine 382 C22H26N2O4 Root R. serpentina Antimalarial
19. Catharanthine 336 C21H24N2O2 Leaves, stem bark, and C. roseus Acethylcholinesterase inhibition
root
20. Vindoline 456 C25H32N2O6 Leaves, stem bark, and C. roseus
root
21. Vinblastine 811 C46H58N4O9 Leaves, stem bark, and C. roseus Chemotherapy
root
22. Vincristine 825 C46H56N4O10 Leaves, stem bark, and C. roseus Chemotherapy
root
S. Kumar and B. Singh
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 515
Various studies available that reported the isolation procedure of different compo-
nents from C. roseus and R. serpentina in detail and identified bisindole alkaloids
(Fig. 3). The various objective behind the old extraction procedures were designated
as selective or distinction extraction, since a weak organic acid (tartaric acid) was
used to bind the stronger bases with the help of acidic proton, allowing the separa-
tion of alkaloids that complex only weakly with the acid, and in this form, are solu-
ble in benzene. The partially deactivated chromatography on alumina with 10%
acetic acid follows, then another form of differential extraction was used. The pH
plays an important role and involved stepwise adjustment of the pH, and extraction
with benzene after each adjustment of pH values. Following this procedure, vincris-
tine and vinrosidine were separated and isolated. Vinblastine and vinleurosine did
not need this procedure, since they could be separated by crystallization rather read-
ily. Vinleurosine was crystallized from methanol, whereas vinblastine could be
crystallized with ethanol but not methanol [17, 20, 49–55].
518 S. Kumar and B. Singh
3 Pharmacological Activities
The crude extracts and isolated MIAs from different parts of C. roseus and R. ser-
pentina have been reported for various pharmacological activities (Fig. 4). The etha-
nolic and methanolic crude extracts of both species have strong pharmacological
bioactivities in both in vivo and in vitro studies. The crude extracts of R. serpentina
and its MIAs have been shown to have effects on the central nervous system and
CVD. C. roseus has been used in chemotherapies. Anthelminthic, antibacterial,
anticancer, antidiabetic, antidiarrheal antihyperlipidemic, antihypertensive, anti-
inflammatory, antimalarial, antimicrobial, antioxidant, antitrypanosomal, cardio-
protective, cholinesterase inhibitory, hepatoprotective, and cytotoxic activities were
reported in different extracts/fractions of plant parts [56–60].
3.1 Chemotherapy
The pharmacological actions of the C. roseus and R. serpentina were reported due
to the presence of indole alkaloids that localized most parts of the plants. Both
C. roseus and R. serpentina are as well one of the most extensively explored
Fig. 4 Different biological activities of the various extracts of C. roseus and R. serpentina
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 519
medicinal herbs. C. roseus is more important and in high demand due to its antican-
cerous bisindole alkaloids, vinblastine and vincristine. Besides, the significance of
C. roseus plant in modern system of medicine has been recognized only after the
coincidence discovery of vinblastine and vincristine which are localized in its
leaves. Currently, four main alkaloids derived from Catharanthus are clinically
accepted – vinblastine, vincristine, vinorelbine, and vindesine; however, only vin-
cristine, vinblastine, and vinorelbine are allowed for clinical use in the United States
[60, 61]. Ajmalicine, ajmaline, reserpine, and serpentine are also isolated from
stems and roots of C. roseus and R. serpentina because of their clinical importance
in the management of high blood pressure and obstructive circulatory problems as
well as enhanced cerebral blood circulation [42, 43]. C. roseus plant parts such as
leaves, stems, and roots also contain serpentine, which is used in the management
of hypertension [46–48]. The discovery of bisindole alkaloids, vincristine and vin-
blastine, isolated from C. roseus symbolizes one of the most imperative introduc-
tions of plant-derived products into cancer chemotherapy. These alkaloids are used
for the treatment of malignant and nonmalignant cancers. Besides chemotherapy,
vincristine and vinblastine are also used in the treatment of thrombocytopenic dis-
orders such as Moschcowitz syndrome (thrombotic thrombocytopenic purpura) and
immune thrombocytopenia (idiopathic thrombocytopenic purpura), and microangi-
opathic hemolytic anemia arises [62, 63]. Even though vincristine and vinblastine
are efficient in platelet and platelet-associated disorders, they are required parts of
the pharmacopoeia that is used for the treatment of malignancy. Vincristine and
vinblastine are also used in a broad spectrum of biochemical applications within the
cells and tissues. The exact mechanism of vincristine and vinblastine within tissue
and cell for cytotoxic action is stated as the interactions with tubulin protein and
disruption of microtubule (spindle fibers) actions, particularly of microtubules com-
prising the mitotic spindle apparatus, directly inducing metaphase arrest. Vinblastine
is also used for the treatment of testicular cancer effectively. Hodgkin lymphomas
and the non-Hodgkin lymphomas are other types of blood cancer that may be treated
with combination regimens that comprise vinblastine. Vinblastine is also useful as
an effective therapy for the treatment of Kaposi’s sarcoma, Alibert-Bazin syndrome
(granuloma fungoides), and breast cancer. Vincristine is also a potent drug of cancer
therapy recommended for the treatment of Hodgkin’s lymphoma and other lympho-
mas, as well as pediatric tumors, for example, nephroblastoma (Wilms tumor) and
embryonal rhabdomyosarcoma. These drugs were initially available in the 1960s;
today, the C. roseus alkaloids are incorporated in each efficient combination chemo-
therapy curriculum, due to their uniqueness in relation to therapeutic action and tox-
icities. Vincristine and vinblastine are unusually imperative in both regimens
(curative and palliative). Some of its alkaloids are approved as antineoplastic agents
for the treatment of leukemia, Hodgkin’s disease, malignant lymphomas, neuroblas-
toma, rhabdomyosarcoma, Wilms’ tumor, and other cancers. Its vasodilating and
memory-enhancing properties have experimentally been showing to alleviate vas-
cular dementia and Alzheimer’s disease [64–66]. The plant also has antihyperten-
sive and antispasmodic properties [67]. The cytotoxic activities of eight bisindole
alkaloids, including catharine, leurosidine, leurosine, vinamidine, etc., have been
520 S. Kumar and B. Singh
reported on MDA-MB-231 cells and were tested by MTT assay. These alkaloids
might induce a marked reduction of the cell viability after 72 h treatment. Among
these, leurosine has exhibited the most potent inhibitory activity, while catharinine
(vinamidine) the weakest activity. It is found that leurosidine showed much weaker
activity than that of vinblastine. The 17-deacetoxyvinamidine has a more potent
activity than catharinine (Vinamidine), implying that the lack of acetoxy at C-17
seems to be an advantage in increasing cytotoxicity. These results provided a clear
indication for further structural modification in bisindole alkaloids [39, 68, 69].
Reserpine (100 ng/ml) significantly enhanced the cytotoxicity of heat-shocked-
activated T cells against both Molt-4 and T98G tumor cells [70]. Yohimbinic acid
and reserpine were found to be the most significant inhibitors against human topoi-
somerase I and II than α-yohimbine, 18-hydroxyepialloyohimbine,
Nb-methylajmaline, Nb-methylisoajmaline, 3-hydroxysarpagine, isorauhimbinic
acid, and reserpinine. Both compounds, yohimbinic acid and reserpine, showed
50% inhibition for topoisomerase I and II at doses of 30 and 20 μM and 20 and
40 μM, respectively, under the same conditions. α-Yohimbine,
18-hydroxyepialloyohimbine, and reserpinine were inactive and had no effect as
inhibitors of topoisomerase I and II. Yohimbinic acid, isorauhimbinic acid, and
reserpine also inhibited human promyelocytic leukemia (HL-60) cell growth, and
reserpine showed an IC50 value of 67 μM. This inhibitory effect of reserpine was
approximately the same as that of dehydrotramentenoic acid which was reported as
an inhibitor of both topo II effect and HL-60 cancer cell growth [71]. Among them,
compound 11-hydroxyburnamine showed promising effects. The crude extract, its
fractions, and its isolated compounds, lupeol, N-methylsarpagine, and spegatrine,
showed inactive cytotoxicity at 50 μg/mL against HeLa cells [72].
3.2 Antihypertensive Therapy
These clinical trials were undertaken to determine whether the hypotensive action
of extracts or tablets of R. serpentina is strictly limited to the period of its adminis-
tration and whether it has prolonged effect. The hypotensive effect of extract of
R. serpentina root (tablets) has been exposed to clinical trial in a series of 55 cases
of important benign hypertension. After treatment for two weeks, the results showed
that the R. serpentina root (tablets) was effective and safe for the treatment of hyper-
tension [73, 74]. Mild to moderate hypertension has been studied via a double-blind
controlled trial using 381 patients in the random distribution. During treatment, the
elevation of the blood pressure effect was reported. A combination of chlorothiazide
(500 mg/kg BWT) and R. serpentina root (100 mg/kg BWT) was more effective if
taken twice daily and did not show any serious side effects. Controlled double-blind
studies showed that compound reserpine acted as a mild antihypertensive agent at
oral doses of 0.8 to 1.5 mg/day BWT; a reduction of blood pressure has also been
reported [75–77]. In a study, the hypertensive effect of a whole-root preparation of
R. serpentina with compound reserpine carried out on 58 patients. It has been
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 521
observed that the whole-root preparation of R. serpentina (400 mg/day) and reser-
pine (0.4 mg/day) showed hypertensive effect without any significant difference.
About 40% of patients who used the preparation showed a satisfactory reduction in
blood pressure without any serious side effects [78]. The extract of the whole root
of R. serpentina or compound reserpine was significantly effective in the treatment
of hypertension with the optimal dose varied from 0–75 to 6–0 mg/kg (serpasil
tablet) and 150–800 mg/kg (raudixin tablet) in 19 ambulatory patients. Sixteen
patients showed reduced blood pressure but with some unavoidable side effects
such as temporary heart failure and weight gain [79]. Thirty-eight patients were
treated with R. serpentina preparations at a dose of 1 g per day for 6 to 20 months.
About 67% of patients showed a reduction of blood pressure (10–20 mm. Hg) con-
sistently after being treated with R. serpentina preparation. The effect of crude root
extract (dosage from 150–450 mg/per day) and compound reserpine (dosage from
0.75–3 mg/ per day) along with addition of apresoline or hexamethonium in a group
of 76 patients with arterial hypertension was studied for time varying between two
and twelve months. The combination of apresoline or hexamethonium with crude
root extract of Rauvolfia showed better results than when given alone. A cases study
was reported for reserpine confirmed as a treatment for constitutional leanness [81].
A comparative study of compounds reserpine and rescinnamine for periods ranging
from 5 to 12 weeks with doses ranging from 1 to 2 mg/ daily in 32 patients showed
that reserpine is more effective than rescinnamine with comparatively minimum
side effects [82].
3.3 Antipsychotic Activity
dose-dependently decreased anxiety from 1.0 to 4.0 mg/kg (p < 0.01) in the light/
dark box. In contrast to extract, the application of reserpine increased anxiety-
related behavior from 0.4 to 0.6 mg/kg using dose-dependently. Therefore, root bark
extract from R. vomitoria could be a better option with great potential as an alterna-
tive antipsychotic agent due to its better behavioral effects with less distortion in
motor coordination as compared to chlorpromazine [85].
3.4 Cardiovascular Diseases
3.5 Psoriasis
4 Conclusions
The growing trend of industrialization and the appearance of many unknown and
tenacious diseases are among the extreme challenges to scientists in near future.
MIAs from C. roseus, R. serpentina have demonstrated novel therapeutic character-
istics as a result of a large number of scientific investigations over the past five
decades. Therefore, vincristine and vinblastine replacing chemical and synthesized
drugs with naturally derived drugs seem highly rationale for the treatment of cancer.
Similarly, various herbal formulations of the R. serpentina and its bioactive
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 523
compounds ajmaline, ajmalicine, and reserpine are used for the treatment of high
blood pressure and hypertension. Additionally, more efforts need to gain insights
into the therapeutic mechanism and toxicological actions of the herb, different
extracts/fractions, and main active MIAs in future studies.
Acknowledgments The authors are thankful to the Principal, MK Govt. Degree College, Ninowa,
Farrukhabad, Uttar Pradesh, India, and Director, CSIR-National Botanical Research Institute
(NBRI), Lucknow, India for encouragement and facilities.
References
18. Svoboda GH, Neuss N, Gorman M et al (1959) Alkaloids of Vinca rosea Linn.(Catharanthus
roseus G. Don.) V. preparation and characterization of alkaloids. J Am Pharm Assoc
48(11):659–666
19. Plummer AJ, Earl A, Schneider JA et al (1954) Pharmacology of Rauwolfia alkaloids, includ-
ing reserpine. Ann N Y Acad Sci 59(1):8–21
20. Itoh A, Kumashiro T, Yamaguchi M et al (2005) Indole alkaloids and other constituents of
Rauvolfia serpentina. J Nat Prod 68(6):848–852
21. Sagi S, Avula B, Wang YH et al (2016) Quantification and characterization of alkaloids from
roots of Rauvolfia serpentina using ultra-high performance liquid chromatography-photo
diode array-mass spectrometry. Anal Bioanal Chem 408(1):177–190
22. Batista CVF, Schripsema J, Verpoorte R (1996) Indole alkaloids from Rauwolfia sellowii.
Phytochemistry 41(3):969–973
23. World Health Organization (2006) The selection and use of essential medicines: report of the
WHO Expert Committee, 2005 (including the 14th Model List of Essential Medicines) (vol
933). World Health Organization
24. Lindenberg M, Kopp S, Dressman JB et al (2004) Classification of orally administered drugs
on the World Health Organization model list of essential medicines according to the biophar-
maceutics classification system. Eur J Pharm Biopharm 58(2):265–278
25. Salam A, Kanukula R, Esam H et al (2019) An application to include blood pressure lowering
drug fixed dose combinations to the model list of essential medicines lists for the treatment of
essential hypertension in adults. World Health Organization, Geneva
26. Arora R, Malhotra P, Mathur AK et al (2010) Anticancer alkaloids of Catharanthus roseus:
transition from traditional to modern medicine. Herbal medicine: a cancer chemopreven-
tive and therapeutic perspective. Jaypee Brothers Medical Publishers Pvt. Ltd, New Delhi,
pp 292–310
27. Ming KJ, Khang GN, Sai CL (2003) Recent advances in traditional plant drugs and orchids.
Acta Pharmacol Sin 24(1):7–21
28. Gao Y, Yu AL, Li GT et al (2015) Hexacyclic monoterpenoid indole alkaloids from Rauvolfia
verticillata. Fitoterapia 107:44–48
29. Gerasimenko I, Sheludko Y, Ma X et al (2002) Heterologous expression of a Rauvolfia cDNA
encoding strictosidine glucosidase, a biosynthetic key to over 2000 monoterpenoid indole
alkaloids. Eur J Biochem 269(8):2204–2213
30. Mehrotra S, Goel MK, Srivastava V et al (2015) Hairy root biotechnology of Rauvolfia ser-
pentina: a potent approach for the production of pharmaceutically important terpenoid indole
alkaloids. Biotechnol Lett 37(2):253–263
31. O'Connor SE, Maresh JJ (2006) Chemistry and biology of monoterpene indole alkaloid bio-
synthesis. Nat Prod Rep 23(4):532–547
32. Stöckigt J, Barleben L, Panjikar S et al (2008) 3D-structure and function of strictosidine syn-
thase–the key enzyme of monoterpenoid indole alkaloid biosynthesis. Plant Physiol Biochem
46(3):340–355
33. Zhu J, Wang M, Wen W et al (2015) Biosynthesis and regulation of terpenoid indole alkaloids
in Catharanthus roseus. Pharmacogn Rev 9(17):24–28
34. Zhu X, Zeng X, Sun C et al (2014) Biosynthetic pathway of terpenoid indole alkaloids in
Catharanthus roseus. Front Med 8(3):285–293
35. Verpoorte R, van der Heijden R, Moreno PR (1997) Biosynthesis of terpenoid indole alka-
loids in Catharanthus roseus cells. In: The alkaloids: chemistry and pharmacology, vol 49.
Academic Press, pp 221–299
36. Zhao J, Zhu WH, Hu Q et al (2001) Effects of light and plant growth regulators on the bio-
synthesis of vindoline and other indole alkaloids in Catharanthus roseus callus cultures. Plant
Growth Regul 33(1):43–49
37. El-Sayed M, Verpoorte R (2007) Catharanthus terpenoid indole alkaloids: biosynthesis and
regulation. Phytochem Rev 6(2):277–305
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 525
58. Abhijit D, De JN (2010) Rauvolfia serpentina (L). Benth ex Kurz-a review. Asian J Plant Sci
9(6):285–298
59. Kumari R, Rathi B, Rani A (2013) Rauvolfia serpentina L. Benth. Ex Kurz.: phytochemical,
pharmacological and therapeutic aspects. Int J Pharm Sci Rev Res 23(2):348–355
60. Neuss N, Neuss MN (1990) Therapeutic use of bisindole alkaloids from catharanthus. In: The
alkaloids: chemistry and pharmacology, vol 37. Academic Press, pp 229–240
61. Nelson RL (1982) The comparative clinical pharmacology and pharmacokinetics of vin-
desine, vincristine, and vinblastine in human patients with cancer. Indian J Med Paediatr
10(2):115–127
62. Pennanen S, Huhtikangas A (1990) Photochemical one-pot synthesis of vinblastine and vin-
cristine. Photochem Photobiol 51(5):515–518
63. Barkat MA, Abul H, Rahman MA (2017) Agricultural, pharmaceutical, and therapeutic inte-
rior of Catharanthus roseus (L.) G. Don. In: Catharanthus roseus. Springer, Cham, pp 71–100
64. Tsuruo T, Iida H, Tsukagoshi S (1981) Overcoming of vincristine resistance in P388 leukemia
in vivo and in vitro through enhanced cytotoxicity of vincristine and vinblastine by verapamil.
Cancer Res 41(5):1967–1972
65. Qu Y, Safonova O, De Luca V (2019) Completion of the canonical pathway for assembly of
anticancer drugs vincristine/vinblastine in Catharanthus roseus. Plant J 97(2):257–266
66. Alam MM, Naeem M, Khan MMA et al (2017) Vincristine and vinblastine anticancer cath-
aranthus alkaloids: pharmacological applications and strategies for yield improvement. In:
Catharanthus roseus. Springer, Cham, pp 277–307
67. Kabesh K, Senthilkumar P, Ragunathan R et al (2015) Phytochemical analysis of Catharanthus
roseus plant extract and its antimicrobial activity. Int J Pure App Biosci 3(2):162–172
68. Wang XD, Li CY, Jiang MM et al (2016) Induction of apoptosis in human leukemia cells
through an intrinsic pathway by cathachunine, a unique alkaloid isolated from Catharanthus
roseus. Phytomedicine 23(6):641–653
69. Parthasarathy R, Shanmuganathan R, Pugazhendhi A (2020) Vinblastine production by the
endophytic fungus Curvularia verruculosa from the leaves of Catharanthus roseus and its
in vitro cytotoxicity against HeLa cell line. Anal Biochem 593:113530
70. Jin GB, Hong T, Inoue S et al (2002) Augmentation of immune cell activity against tumor cells
by Rauwolfia radix. J Ethnopharmacol 81(3):365–372
71. Chen Q, Lu X, Guo X (2017) Metabolomics characterization of two Apocynaceae plants,
Catharanthus roseus and Vinca minor, using GC-MS and LC-MS methods in combination.
Molecules 22(6):997
72. Tlhapi DB, Ramaite ID, Anokwuru CP et al (2020) In vitro studies on antioxidant and anti-
parasitic activities of compounds isolated from Rauvolfia caffra Sond. Molecules 25(17):3781
73. Vakil RJ (1949) A clinical trial of Rauvolfia serpentina in essential hypertension. Br Heart J
11(4):350–355
74. Vakil RJ (1955) Rauvolfia serpentina in the treatment of high blood pressure: a review of the
literature. Circulation 12(2):220–229
75. Smith WM, Damato AN, Galluzzi NJ et al (1964) The evaluation of antihypertensive ther-
apy cooperative clinical trial method: I. Double-blind control comparison of chlorothiazide,
Rauvolfia serpentina, and hydralazine. Ann Intern Med 61(5_Part_1):829–846
76. Smith WM, Bachman B, Galante JG et al (1966) Co-operative clinical trial of alpha-
methyldopa: III. Double-blind control comparison of alpha-methyldopa and Chlorothiazide,
and Chlorothiazide and Rauwolfia. Ann Intern Med 65(4):657–671
77. Smith WM, Thurm RH, Bromer L (1969) Comparative evaluation of Rauwolfia whole root and
reserpine. Clin Pharmacol Ther 10(3):338–343
78. Healy D, Savage M (1998) Reserpine exhumed. Br J Psychiatry 172(5):376–378
79. McGregor M, Segel N (1955) The Rauwolfia alkaloids in the treatment of hypertension. Br
Heart J 17(3):391–396
80. Genest J, Adamkiewicz L, Robillard R et al (1955) Clinical uses of Rauwolfia: I. In arterial
hypertension. Can Med Assoc J 72(7):483–489
Phytochemistry and Pharmacology of Catharanthus roseus (L.) G. Don… 527
81. Lemieux G, Davignon A, Genest J (1956) Depressive states during Rauwolfia therapy for arte-
rial hypertension: a report of 30 cases. Can Med Assoc J 74(7):522
82. Bisong SA, Brown RE, Osim EE (2013) Comparative extrapyramidal effects of Rauwolfia
vomitoria, chlorpromazine and reserpine in mice. J Nat Med 67(1):107–112
83. Gupta S, Khanna VK, Maurya A et al (2012) Bioactivity guided isolation of antipsychotic
constituents from the leaves of Rauwolfia tetraphylla L. Fitoterapia 83(6):1092–1099
84. Bisong SA, Brown R, Osim EE (2010) Comparative effects of Rauwolfia vomitoria and chlor-
promazine on locomotor behaviour and anxiety in mice. J Ethnopharmacol 132(1):334–339
85. Surendran S, Raju R, Prasannan P et al (2021) A comprehensive review on ethnobotany, phy-
tochemistry and pharmacology of Rauvolfia L.(Apocynaceae). The Bot Rev:1–66
Resilience Activity of Glycyrrhiza glabra
in Relation to Cancer: Chemistry
and Mechanism
1 Introduction
From ancient times, plants are considered major sources of medicine and are used
in the prevention and elimination of diseases. Today across the world, there has
been enormous demand for herbal medicines because of their safety, efficacy, and
lesser toxicity in comparison to synthetic drugs [1]. Glycyrrhiza glabra (Family:
Fabaceae), also known as licorice derived from the Greek words glykos (sweet) and
rhiza (root), is one of the most popular medicinal plants used in traditional medicine
to treat varieties of diseases [2–7]. Glycyrrhiza glabra has long history of folk and
traditional medicines and it is one of the oldest known botanical medicinal herbs.
About 90% of G. glabra is used as flavouring agents in tobacco products, chewing
tobacco, pipe tobacco, and American blend cigarettes [8, 9]. G. glabra-based fla-
vouring agents are added in soft drinks, herbal teas, candies, chewing gum, tooth-
paste, and herbal remedies for stomach problems and cough [7, 10]. Annual
consumption of licorice is about 1.5 kg/person worldwide [11].
G. glabra is known for various pharmacological, anticarcinogenic, antitussive,
antimicrobial, anticoagulant, antiviral, antiulcer, antioxidant, antiinflammation,
antidiabetic, immunomodulatory, hepatoprotective, and antiatherogenic activities
(Fig. 1) [6, 12–32].
Researchers have reported that various secondary metabolites are present in
G. glabra. It is reported that G. glabra contains more than 300 flavonoids and 20
N. Dhingra (*)
Department of Agriculture, Medi-Caps University, Indore, Madhya Pradesh, India
S. Sharma · P. Kumari
Department of Biology, College of Arts and Sciences, Georgia State University,
Atlanta, GA, USA
A. Kar
School of Life Sciences, Devi Ahilya University, Indore, Madhya Pradesh, India
2 Phytochemistry/Chemistry
Many bioactive components have been isolated from the roots of Glycyrrhiza gla-
bra, using different solvents such as chloroform, petroleum ether, n-butanol, metha-
nolic and water [34]. Qualitative analysis of these extracts shows the presence of a
wide range of bioactive components, categorized into different groups, including
saponin, mucilage (from rhizome), terpenoids, amino acids, flavonoids, phenols,
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 531
and various other substances [7, 41]. Flavonoid-rich fraction includes liquirtin,
liquiritigenin, glucoliquiritin apioside, coumarin-GU-12, etc., while four new
isoprenoids-phenolic substitutes such as isoangustone A, semilicoisoflavone B, lic-
oriphenone, and 1-methoxyficifolinol were reported [42, 43]. Some of the volatile
components identified include pentanol, tetramethyl pyrazine, hexanol,
terpinene-4-ol, linalool oxide A and B, geraniol, and α-terpineol. Triterpenoid sapo-
nin glycyrrhizin, a key ingredient in Glycyrrhiza glabra root extracts, is known for
its anti-cancerous sweetener (60 times more potent than regular cane sugar) activity
[42]. However, methanolic extract showed a strong bactericidal effect against both
gram-positive and gram-negative bacteria, with most robust activity against
Staphylococcus aureus with the zone of inhibition 22 mm [43].
532 N. Dhingra et al.
2.1 Glycyrrhizin
Representing 10% of the root dry weight, glycyrrhizin (dry weight) makes up most
of the Glycyrrhiza glabra plant root. Chemically, glycyrrhizin is a mixture of potas-
sium, calcium, and magnesium salts of glycyrrhizin acid (2–25%) [7]. Structurally,
glycyrrhizin consists of glycyrrhizin acid that is composed of a hydrophobic frag-
ment, two molecules of glucuronic acid, and a hydrophilic part [44]. Glycyrrhizin,
a triterpenoid of aglycone, and glycyrrhetic acid bonded with a glucuronic acid
disaccharide can exist as stereoisomers of 18α and 18β. Licorice extract was used to
obtain ammoniated salt of glycyrrhizin, which is further used in the food industry
for flavour [45]. An analog of glycyrrhizin acid, carbenoxolone, is used to treat
digestive disorders, such as peptic ulcers [45].
2.2 Liquorice
3 Pharmacological Properties
Glycyrrhiza glabra extracts prepared in various solvents have been used to identify
the pharmacological potential of the plant. In vivo and in vitro experiments using
the root and leave extracts have shown potent anti-inflammatory, antimicrobial,
antiproliferative, and cytotoxic activities. Nonetheless, the biological activity varies
following the geographical distribution of the plants, extraction methodology, dry-
ing, and harvesting methods.
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 533
Glycyrrhiza glabra has been primarily studied because of its antioxidant potential.
Phenolic and flavonoid extracts were attributed to their antioxidant effect, while
their derivatives enhance antioxidant potential. Glabridin, hispaglabridin A, and
30-hydroxy-40-methylglabridin are the major phenolic composition in ethanolic
extract, conferring the antioxidant potential through metal chelating, donation of
hydrogens, reducing ability, and scavenging assay. Phenolic compounds protect
against oxLDL, lipid peroxidation (microsomal and mitochondrial), and oxidative
stress, making them an ideal candidate for cardiovascular research, in addition to
known health benefits [48]. Retrochalcone, derivative from Glycyrrhiza glabra,
exerts strong antioxidant and protect RBC from oxidative haemolysis. Licochalcone
C supresses superoxide radicals and promotes nitric oxide synthesis [49]. Methanolic
extract mixed with DPPH (1,1-diphenyl-2-picrylhydrazyl) showed scavenging
potential of 67.2% at a concentration of 500ug/ml [50].
Licorice extract from Glycyrrhiza exhibits anti-inflammatory activity through
inhibition of nitric oxide, IL-6, PGE2 (prostaglandin E2) in LPS (lipopolysaccha-
ride) induced macrophages [51]. Mueller et al. (2010) confirmed that Glycyrrhiza of
root extract (0.5–0.2 mg/ml) promotes anti-inflammatory activity by suppressing
COX2, iNOS, TNFa, IL1b, and IL8 [52, 53]. Oral administration of Male albino rats
(Sprague Dawley strain) with Glycyrrhiza glabra root extract (200-800 mg/kg BW)
was done for 4-weeks. Significant improvements in total cholesterol and triglycer-
ides decrease serum liver enzyme (AST and ALT), inducing potent anti-inflammatory
and anti ulcerative effects [54]. Mechanistically, digested Glycyrrhiza is broken
down into various subunits, part of which act similar to hydrochortison (steroid
hormone) to inhibit PLA2 (phospholipase) and platerlet aggregations [55].
Glycyrrhizin, a potent anti-inflammatory compound isolated from the Glycyrrhiza,
is known to increase blood coagulation time and plasma recalcification, therefore
categorized as the first plant-based thrombin inhibitor without affecting the collagen
or platelet aggregation factors [16, 56]. Collectively, in vitro and in vivo analyses
suggest that Glycyrrhiza glabra is a potent antioxidant and anti-inflammatory as
compared to known vitamins [57].
At the current pandemic, viral disease has been a significant concern around the
globe. According to Baltina et al. (2003) glycoprotein derivatives of Glycyrrhizic
Acid have been shown to possess immunomodulatory activity in mice. Intraperitoneal
injection of Glycyrrhizic Acid (2 mg/kg) derivatives with methyl esters of amino
acids stimulated the immune system in stressed mice. In combination with zinc
(45 mg/kg), Glycyrrhiza root extracts (1.5 g/kg BW) protect against systemic ana-
phylactic reactions and promote immunomodulation in rodents [58]. However,
534 N. Dhingra et al.
Free radicals generated during the cellular process promote neurological disorders,
including memory loss and depression. The antioxidant effect of licorice (flavonoid)
is attributed to its ability to neutralize free radicles to improve neurological disor-
ders [42, 66]. 500 mg tablets prepared from grounded roots (crude) of Glycyrrhiza
glabra were used to evaluate intelligence and memory function in male adult kids
(n = 123) aged 13-14 years [67]. Non-verbal intelligence test (NVIT) among the test
and placebo group showed that oral consumption of 500 mg tablets for 6 months
improved student intelligance in the test group compared with the placebo group
[67]. The drug treatment showed overall improvement in general intelligence rather
than short-term intelligence improvement [67]. In a similar experiment on rodents
(3 months old, Wistar albino rats), Chakravarthi et al. (2012) used Elevated plus-
maze and Morris water maze tests to evaluate the effect of Glycyrrhiza glabra root
extract on memory. Rats were fed with variable concentration of root extracts (aque-
ous) for 2 weeks followed by evaluation of cognitive functions. 150 mg/kg dose
effectively enhanced cognitive ability in rats; additionally, a liner dosage-dependent
improvement on learning and memory was also reported [68]. Additionally, the
principle isoflavone glabridin exhibits strong sedative and hypnotic effects through
the inhibition in dorsal raphe neurons by the activation of γ-amino butyric acid-A
(GABAA) receptor [69]. Isoliquiritigenin, a key phytocompound of glycyrrhiza,
forms a significant portion of Japanese Kampo Medicine Yokukansan, promoting
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 535
In vitro and In vivo experiments using Glycyrrhiza extracts have shown promising
results. Some of the primary compounds that have been tested against anti-cancerous
activity include glabridin, licochalcone E, licochalcone A, 18β-glycyrrhetinic, etc.
The principal isoflavonoid compound, glabridin at a concentration of 0-100uM
(in vitro), or 0-20 mg/kg (in vivo), can effectively supress cancerous properties in
breast cancer stem cells and can induce apoptosis in hepatic cell lines such as Huh7,
HepG2, Sk-Hep-1 in vitro and in vivo. Glabridin enhances the expression of
microRNA-148 through DNA methylation, followed by the binding of
microRNA-148 at 3’UTR of SMAD2 to supress TGFb-induced stemness in breast
cancer cells (microRNA-148/TGFb-SMAD2 signalling) [72, 73]. Licochalcone E
and licochalcone A induce apoptosis in pharyngeal squamous cells (FaDu) and gas-
tric cell lines in a caspase and mitochondrial-dependent manner, like that of cell
death in human oral keratinocytes (hNOKs) cell line [74, 75]. FaDu cells when
treated with 50uM of licochalcone E for 24 hr. resulted in increased expression of
pro-apoptotic factors such as caspase 8, BAX, Bcl-2, caspase 9, cleaved caspase3,
and poly (ADP-ribose) polymerase [74].
Scientific literature suggests that 18β-glycyrrhetinic and glycyrrhizic acids effec-
tively supress the growth of cervix and uterine tumours through activation of pro-
apoptotic Bax, cytochrome C, caspase-3, and by disruption of mitochondrial
membrane permeability [76]. In hepatic tissue, 18β-glycyrrhetinic inhibited prolif-
eration and migration of cancerous cells while promoting a healthy tumour micro-
environment by suppressing NF-kB, iNOS, and COX2 activity [41, 77]. In breast
cancer cell lines and mammary epithelial cells, 100uM of 18β-glycyrrhetinic inacti-
vates and inhibits nuclear translocation of FOXO3a, thereby inhibiting the Akt
kinase activity mediated by caspase-9 [78]. In HL-60 (human leukaemia), glycyr-
rhetinic induced cell death through intrinsic and extrinsic pathways [79]. Methanolic
extract of Glycyrrhiza possesses cytotoxic, antimicrobial, and antioxidant activity.
Brine shrimp lethality bioassay showed cytotoxicity with LC50 of 0.77ug/ml [43].
Treatment of colorectal cancer cell lines such as LoVo, SW480, and SW620 with 18
β-glycyrrhetinic acids suppressed proliferation and migration in a dosage-dependent
manner in vitro and in vivo. Annexin V-FITC staining showed reduced survivin
expression and increased PARP expression, along with suppression of p-PI3K,
p-AKT, p-STAT3, p-JNK p-p38, and p-NF-κB p65, thereby regulating apoptosis,
invasion, migration, and proliferation of colorectal cancer [80].
Hydromethanolic Glycyrrhiza extract protected against cyclophosphamide-
induced mutagenicity in bone marrow cells of Swiss albino mice. Intra-peritoneally
536 N. Dhingra et al.
5 Mechanism
Glycyrrhizin (GL), a triterpene glycoside (saponin), is the chief sweet tasting con-
stituent of Glycyrrhiza glabra (liquorice) root. It is one of the major components of
licorice, also known as glycyrrhizin acid. Various mechanisms of GL have been
reported such as reduction in the expression of proliferation of proliferative cell
nuclear antigen (PCNA), exhibition of fewer TUNEL (terminal deoxynucleotidyl
transferase-mediated dUTP nick end labelling)-positive cells, and increased
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 537
5.1 Isoliquiritigenin
Isoliquiritigenin (ISL) with a chalcone structure is one of the most important bioac-
tive components derived from licorice root [82, 83, 86]. ISL is known to possess
antitumour activity by various mechanisms which include cell proliferation, angio-
genesis, invasion, epigenesis, and autophagy in different types of cancer [82, 83].
Various researchers reported that ISL significantly acts on various breast cancer
cells by enhancing the expression of presenilin2 mRNA level, WIF1, HIF-1α pro-
teasome degradation, cleaved caspase-3 & 9, RECK, PTEN, Bax, MMP-7, PIAS3,
and many more. Similarly, it suppresses the expression of estrogen receptor α,
DNMT1, β-catenin, c-myc, VEGF/VEGFR-2, and many more [87, 88]. In colon
cancer, ISL inhibits AMPK and AKT/mTOR pathway and mediates HIF-1α which
inhibits glycolysis and lactate degeneration. ISL downregulates antiapoptotic pro-
teins Bcl-2 and Bcl-x(L), arrested in G2, and thus inhibits tumour growth. It also
induces apoptosis by reducing PGE2 and nitric oxide (NO) in mouse and human
colon carcinoma cells. Studies have shown that ISL induces apoptosis and cytotox-
icity by suppressing PI3K/AKT/mTOR pathway, increasing Beclin-1, by regulating
calcium and Delta psi(m)-dependent pathways, inhibiting m-TOR, by increasing
p21Cip1/Waf, reducing Bcl-2, cdk 2/4, and E2F, elevation of FAS ligand in many
cancerous cells including ovary, prostate, gastric, osteosarcoma, glioma, bladder,
etc. [82, 83, 86–88].
5.2 Isoangustone A
D1 and cyclin E protein. They also reported that IAA suppressed the phosphoryla-
tion of PI3k, MKK4, MKK7, GSK3β, Akt, and JNK1/2 [89]. In one of the studies,
it has been reported that IAA induces apoptosis in human colorectal adenocarci-
noma cells by disrupting mitochondrial functions. IAA induces apoptosis by disrup-
tion of mitochondrial structures, dissipation of mitochondrial membrane potential,
and release of cytochrome C. It also inhibited the phosphorylation of Akt and acti-
vated AMPK in colorectal cancer. Lee et al. (2013) reported that IAA suppresses the
growth of human prostate cancer cell by supressing the kinase activity of CDK2 and
mTOR and thus inhibits the phosphorylation of Akt and p27 [81–117] (Table 1).
5.3 Licochalcone
Licochalcones (LC) which are found in and extracted from the roots of Glycyrrhiza
species G. glabra are derivatives of phenol chalconoid and are known to possess
anti-cancer property. There has been reported that LC-A increase reactive oxygen
species (ROS), human gastric cancer cells, and thus cause apoptosis. LC-A is
reported to activate ERK, JNK, and p38 MAPK in BGC-823 cells which leads to the
apoptosis of cells. Lee et al. (2008) reported that LC-A significantly inhibited the
size of solid tumours in CT-26 cells-inoculated Balb/c mice. Various researchers
reported the effect of LC-A in various cancers including bladder, colorectal, colon,
human oesophageal squamous carcinoma, etc., by diversified mechanism, viz.,
decreased expression of cyclin A, cyclin B, MDM2, and many more. Oh et al.
(2019) reported that LC-B directly binds to the ATP binding pocket of EGFR and
MET kinase and induces apoptosis in human non-small-cell lung cancer cells. LC-C
is known to induce apoptosis in human oral squamous cell carcinoma cells by regu-
lating JAK2/STAT3 signalling pathways. Kwak et al. (2020) reported that LC-C
induced cycle arrest by increase in the expression p21 and p27 and by decreased
cyclin D1. LC-C induced the phosphorylation JNK, c-Jun, and p38 protein [74, 75].
6 Conclusion
Glycyrrhiza glabra is considered one of the most popular medicinal plants used in
traditional medicine. It is used in various industrial products, viz. candies, chewing
gum, herbal teas, tobacco products, etc. It consists of diversified pharmacological
activities including cancer. It is known to have enormous bioactive components
including flavonoids, tannins, saponins, terpenoids glycyrrhetic acid, glycyrrhizin,
glabridin, etc. Different extracts and bioactive components of G. glabra are known
to possess anti-cancer activity on different types of cancers. Various mechanisms
are proposed for their anti-cancer activity like chromatin condensation, DNA frag-
mentation, apoptosis, and down- and up-regulation of many proteins. G. glabra
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 539
possesses strong agent for new molecules which could have immense medicinal
properties and might be a therapeutic target for various cancer.
References
23. Salvi M et al (2003) Glycyrrhetinic acid-induced permeability transition in rat liver mito-
chondria. Biochem Pharmacol 66(12):2375–2379
24. Fiore C et al (2004) On the mechanism of mitochondrial permeability transition induction by
glycyrrhetinic acid. Biochim Biophysi Acta 1658(3):195–201
25. Watanabe M et al (2002) Identification of licocoumarone as an apoptosis-inducing compo-
nent in licorice. Biol Pharm Bull 25(10):1388–1390
26. Sharma V, Agrawal R, Shrivastava VK (2014) Assessment of median lethal dose and anti-
mutagenic effects of Glycyrrhiza glabra root extract against chemically induced micronu-
cleus formation in Swiss albino mice. Int J Basic Clin Pharmacol 3:292–297
27. Acharya S et al (1993) A preliminary open trial on interferon stimulator (SNMC) derived from
Glycyrrhiza glabra in the treatment of subacute hepatic failure. Indian J Med Res 98:69–74
28. Sato H et al (1996) Therapeutic basis of glycyrrhizin on chronic hepatitis B. Antivir Res
30(2–3):171–177
29. Van Rossum T et al (1998) Glycyrrhizin as a potential treatment for chronic hepatitis
C. Aliment Pharmacol Ther 12(3):199–205
30. Jeong HG et al (2002) Hepatoprotective effects of 18β-glycyrrhetinic acid on carbon
tetrachloride-induced liver injury: inhibition of cytochrome P450 2E1 expression. Pharmacol
Res 46(3):221–227
31. Chan H-t, Chan C, Ho JW (2003) Inhibition of glycyrrhizic acid on aflatoxin B1-induced
cytotoxicity in hepatoma cells. Toxicology 188(2–3):211–217
32. Sharma V, Agrawal R (2014) In vivo antioxidant and hepatoprotective potential of Glycyrrhiza
glabra extract on carbon tetra chloride (CCl4) induced oxidative-stress mediated hepatotoxic-
ity. Int J Res Med Sci 2(1):314–320
33. Takii H et al (2001) Antidiabetic effect of glycyrrhizin in genetically diabetic KK-ay mice.
Biol Pharm Bull 24(5):484–487
34. Husain A et al (2015) Quantitative analysis of total phenolic, flavonoid contents and HPTLC
fingerprinting for standardization of Glycyrrhiza glabra Linn. Roots. Herb Med 1(1):1–9
35. Bradley PR (1992) British herbal compendium. Volume 1. A handbook of scientific informa-
tion on widely used plant drugs. Companion to Volume 1 of the British Herbal Pharmacopoeia.
British Herbal Medicine Association
36. Isbrucker R, Burdock G (2006) Risk and safety assessment on the consumption of Licorice
root (Glycyrrhiza sp.), its extract and powder as a food ingredient, with emphasis on the phar-
macology and toxicology of glycyrrhizin. Regul Toxicol Pharmacol 46(3):167–192
37. Capasso F, De Pasquale R, Grandolini G (2011) Farmacognosia: Botanica, chimica e farma-
cologia delle piante medicinali. Springer Science & Business Media
38. Li J-R, Wang Y-Q, Deng Z-Z (2005) Note: two new compounds from Glycyrrhiza glabra. J
Asian Nat Prod Res 7(4):677–680
39. Wren RC, Wren RW (1972) Potter’s new cyclopaedia of medicinal herbs and preparations.
Harper & Row
40. Kinoshita T, Tamura Y, Mizutani K (2005) The isolation and structure elucidation of minor
isoflavonoids from licorice of Glycyrrhiza glabra origin. Chem Pharm Bull 53(7):847–849
41. Hasan MK et al (2021) Phytochemistry, pharmacological activity, and potential health ben-
efits of Glycyrrhiza glabra. Heliyon 7:e07240
42. Yu J-Y et al (2015) Anti-inflammatory activities of licorice extract and its active com-
pounds, glycyrrhizic acid, liquiritin and liquiritigenin, in BV2 cells and mice liver. Molecules
20(7):13041–13054
43. Sultana S et al (2010) Antimicrobial, cytotoxic and antioxidant activity of methanolic extract
of Glycyrrhiza glabra. Agric Biol J N Am 1(5):957–960
44. Obolentseva G et al (1999) Pharmacological and therapeutic properties of licorice prepara-
tions (a review). Pharm Chem J 33(8):427–434
45. Experts, U.S.P.C.C.o. and U.S.P.C.F.I.E. Committee, Food Chemicals Codex (2010) US
Pharmacopeia Conv
542 N. Dhingra et al.
46. Simmler C, Pauli GF, Chen S-N (2013) Phytochemistry and biological properties of gla-
bridin. Fitoterapia 90:160–184
47. Fukui H, Goto K, Tabata M (1988) Two antimicrobial flavanones from the leaves of
Glycyrrhiza glabra. Chem Pharm Bull 36(10):4174–4176
48. Sharma V, Katiyar A, Agrawal R (2018) Glycyrrhiza glabra: chemistry and pharmacological
activity. Sweeteners:87
49. Franceschelli S et al (2011) Licocalchone-C extracted from Glycyrrhiza glabra inhibits
lipopolysaccharide-interferon-γ inflammation by improving antioxidant conditions and regu-
lating inducible nitric oxide synthase expression. Molecules 16(7):5720–5734
50. Chopra P et al (2013) Antimicrobial and antioxidant activities of methanol extract roots of
Glycyrrhiza glabra and HPLC analysis. Int J Pharm Pharmacol Sci 5(2):157–160
51. Fu Y et al (2013) Antioxidant and anti-inflammatory activities of six flavonoids separated
from licorice. Food Chem 141(2):1063–1071
52. Bodet C et al (2008) A licorice extract reduces lipopolysaccharide-induced proinflammatory
cytokine secretion by macrophages and whole blood. J Periodontol 79(9):1752–1761
53. Mueller M, Hobiger S, Jungbauer A (2010) Anti-inflammatory activity of extracts from
fruits, herbs and spices. Food Chem 122(4):987–996
54. Shalaby MA et al (2004) Some effects of Glycyrrhiza glabra (liquorice) roots extract on male
rats. Egy J Nat Toxins 1:83–94
55. Harwansh RK, Patra KC, Pareta SK, Singh J, Biswas R (2011) Pharmacological studies on
Glycyrrhiza glabra: a review. Pharmacologyonline 2:1032–1038
56. Bhuiyan MYH et al (2015) Standardization, quality control and pharmacological review on
Glycyrrhiza glabra L. A potential medicinal herb in Unani and Ayurvedic systems of medi-
cine. Hamdard Med 58:45–72
57. Damle M (2014) Glycyrrhiza glabra (Liquorice)-a potent medicinal herb. Int J Herb Med
2(2):132–136
58. Mazumder PM et al (2012) Evaluation of immunomodulatory activity of Glycyrhiza glabra L
roots in combination with zing. Asian Pac J Trop Biomed 2(1):S15–S20
59. Vikhe G et al (2013) In vitro effect of G. glabra and T. Cordifolia plant extracts on phagocy-
tosis by human neutrophils. Pravara Med Rev 5(2)
60. Laconi S, Madeddu MA, Pompei R (2014) Autophagy activation and antiviral activity by a
licorice triterpene. Phytother Res 28(12):1890–1892
61. Michaelis M et al (2010) Glycyrrhizin inhibits highly pathogenic H5N1 influenza a virus-
induced pro-inflammatory cytokine and chemokine expression in human macrophages. Med
Microbiol Immunol 199(4):291–297
62. Baltina L (2003) Chemical modification of glycyrrhizic acid as a route to new bioactive com-
pounds for medicine. Curr Med Chem 10(2):155–171
63. Soufy H et al (2012) Antiviral and immune stimulant activities of glycyrrhizin against duck
hepatitis virus. Afr J Tradit Complement Altern Med 9(3):389–395
64. Fukuchi K et al (2016) Antiviral and antitumor activity of licorice root extracts. In Vivo
30(6):777–785
65. Cinatl J et al (2003) Glycyrrhizin, an active component of liquorice roots, and replication of
SARS-associated coronavirus. Lancet 361(9374):2045–2046
66. Kim HJ et al (2012) Antioxidant activities of licorice-derived prenylflavonoids. Nutr Res
Pract 6(6):491–498
67. Teltumbde A et al. (2013) Effect of Yashtimadhu (Glycyrrhiza glabra) on intelligence and
memory function in male adolescents. Sch J App Med Sci 1(2):90–95
68. Chakravarthi KK, Avadhani R, Narayan RS (2012) Effect of Glycyrrhiza glabra root extract
on learning and memory in wistar albino rats. Drug Invent Today 4(7):387–390
69. Jin Z et al (2013) Potentiating effect of glabridin on GABAA receptor-mediated responses in
dorsal raphe neurons. Planta Med 79(15):1408–1412
70. Ikarashi Y, Mizoguchi K (2016) Neuropharmacological efficacy of the traditional Japanese
Kampo medicine yokukansan and its active ingredients. Pharmacol Ther 166:84–95
Resilience Activity of Glycyrrhiza glabra in Relation to Cancer: Chemistry… 543
71. Danysz W, Parsons CG (2003) The NMDA receptor antagonist memantine as a symptom-
atological and neuroprotective treatment for Alzheimer’s disease: preclinical evidence. Int J
Geriatr Psychiatry 18(S1):S23–S32
72. Jiang F et al (2016) Glabridin inhibits cancer stem cell-like properties of human breast cancer
cells: an epigenetic regulation of miR-148a/SMAd2 signaling. Mol Carcinog 55(5):929–940
73. Hsieh M-J et al (2016) Glabridin induces apoptosis and autophagy through JNK1/2 pathway
in human hepatoma cells. Phytomedicine 23(4):359–366
74. Yu SJ et al (2017) Licochalcone-E induces caspase-dependent death of human pharyngeal
squamous carcinoma cells through the extrinsic and intrinsic apoptotic signaling pathways.
Oncol Lett 13(5):3662–3668
75. Xiao X-Y et al (2011) Licochalcone a inhibits growth of gastric cancer cells by arresting cell
cycle progression and inducing apoptosis. Cancer Lett 302(1):69–75
76. Lee CS et al (2008) 18β-Glycyrrhetinic acid induces apoptotic cell death in SiHa cells
and exhibits a synergistic effect against antibiotic anti-cancer drug toxicity. Life Sci
83(13–14):481–489
77. Hasan SK et al (2016) Chemopreventive effect of 18β-glycyrrhetinic acid via modulation of
inflammatory markers and induction of apoptosis in human hepatoma cell line (HepG2). Mol
Cell Biochem 416(1–2):169–177
78. Sharma G et al (2012) 18β-glycyrrhetinic acid induces apoptosis through modulation of Akt/
FOXO3a/Bim pathway in human breast cancer MCF-7 cells. J Cell Physiol 227(5):1923–1931
79. Huang Y-C et al (2016) 18α-glycyrrhetinic acid induces apoptosis of HL-60 human leu-
kemia cells through caspases-and mitochondria-dependent signaling pathways. Molecules
21(7):872
80. Wang S et al (2017) 18 β-glycyrrhetinic acid exhibits potent antitumor effects against colorec-
tal cancer via inhibition of cell proliferation and migration. Int J Oncol 51(2):615–624
81. Deng Q-P et al (2017) Effects of glycyrrhizin in a mouse model of lung adenocarcinoma. Cell
Physiol Biochem 41(4):1383–1392
82. Hsu Y-L, Kuo P-L, Lin C-C (2005) Isoliquiritigenin induces apoptosis and cell cycle arrest
through p53-dependent pathway in Hep G2 cells. Life Sci 77(3):279–292
83. Yamazaki S et al (2002) Isoliquiritigenin suppresses pulmonary metastasis of mouse renal
cell carcinoma. Cancer Lett 183(1):23–30
84. Farooqui A et al (2018) Glycyrrhizin induces reactive oxygen species-dependent apoptosis
and cell cycle arrest at G0/G1 in HPV18+ human cervical cancer HeLa cell line. Biomed
Pharmacother 97:752–764
85. Kabe Y et al (2021) Glycyrrhizin derivatives suppress cancer chemoresistance by inhibiting
progesterone receptor membrane component 1. Cancers (Basel) 13(13):3265
86. Wang K-L, Yu Y-C, Hsia S-M (2021) Perspectives on the role of Isoliquiritigenin in cancer.
Cancers (Basel) 13(1):115
87. Maggiolini M et al (2002) Estrogenic and antiproliferative activities of isoliquiritigenin in
MCF7 breast cancer cells. J Steroid Biochem Mol Biol 82(4–5):315–322
88. Wang N et al (2015) Dietary compound isoliquiritigenin prevents mammary carcinogenesis
by inhibiting breast cancer stem cells through WIF1 demethylation. Oncotarget 6(12):9854
89. Song NR et al (2013) Isoangustone A, a novel licorice compound, inhibits cell proliferation by
targeting PI3K, MKK4, and MKK7 in human melanoma. Cancer Prev Res 6(12):1293–1303
90. Lee E et al (2013) CDK2 and mTOR are direct molecular targets of isoangustone A in the
suppression of human prostate cancer cell growth. Toxicol Appl Pharmacol 272(1):12–20
91. Huang R-Y et al (2014) Glycyrrhizin suppresses lung adenocarcinoma cell growth through
inhibition of thromboxane synthase. Cell Physiol Biochem 33(2):375–388
92. Zhu J et al (2021) Licorice extract inhibits growth of non-small cell lung cancer by down-
regulating CDK4-cyclin D1 complex and increasing CD8+ T cell infiltration. Cancer Cell
Int 21(1):1–18
93. Wu X et al (2018) Glycyrrhizin suppresses the growth of human NSCLC cell line HCC827
by downregulating HMGB1 level. Biomed Res Int 2018
544 N. Dhingra et al.
1 Introduction
Traditional medicine was purely based on plant and plant products. The vast variety
of medicinal plants were explored by our ancestors in ancient times itself where
modern technology had not even named them. Each plant has its medicinal property
which can be used as a potential drug against diseases. With the help of modern
sophisticated technologies, the underexplored and unknown pharmaceutical activi-
ties of plants were revealed nowadays. Likewise, the genus Strobilanthes were used
in traditional medicine from time immemorial; still, the complete picture in respect
of the bioactivities and medicinal quality of the genus is not discovered so far. The
bioactivities, phytochemicals revealed, and the chromosome reports of the species
in the genus so far discovered are discussed here.
Chinese, it is called Hei Mian Jiang Jun which means Black-faced General. Chinese
drug ‘Shengma’ is a combination of Serratula and Strobilanthes [4].
Ban-lan-gen is used to describe root tissues of several indistinguishable plant
species in China which are used as medicine. Indigo, indirubin, and tryptanthrin are
the key compounds of Ban-len-gen [5]. S. cusia is one of them which is widely used
in the textile industry due to rich indigo. Transcriptomic analysis revealed that the
genes encoding the indigo biosynthesis were monooxygenase, uridine diphosphate-
glycosyl transferase, and b-glucosidase. The genetic information of dye-yielding
plants can increase the application of genetic engineering in industries [6]. S. ban-
tonensis is called ‘purple Ban-len-gen’ due to its similarity in root with S. cusia.
This herb is used against influenza virus infection in China and Vietnam [7].
Indigo Naturalis is used against skin psoriasis and nail psoriasis. Nail psoriasis
includes nail dystrophy, thickening, loss of lustre, raising, color changes, and fria-
bility. It is a powder extracted from the branches and leaves of indigo-producing
plants by fermenting in fresh water for several days of decomposition. Ethyl acetate
extract of Indigo Naturalis isolated from S. formosanus inhibits gram-positive bac-
teria, mildly inhibits non-dermatophytic onychomycosis pathogen, and has no effect
on dermatophytes [8].
In ancient times, the tribals used roots of S. callosus for the treatment of inflam-
matory disorders and its stem bark in formulations for painful and ineffectual
attempts to urinate or defecate [9]. The roots of S. cilliatus were used for the treat-
ment of rheumatalgia, limping, chest congestion, strangury, fever, leucoderma, skin
diseases, and inflammation [10]. Its stem is widely used for whooping cough, bron-
chitis, dropsy, leprosy, and pruritus [11]. Indigo Naturalis is used from earlier times
as textile dye and paint pigment. Modern research has revealed that Indigo Naturalis
is used as an antipyretic, anti-inflammatory, antiviral, antimicrobial, anticancer, and
detoxicant agents [12]. Strobilanthes is a good source of Indigo Naturalis, which is
used in Chinese medicine also [8].
S. cilliatus leaves are rich in potassium. Lupeol derived from its root is used to
treat skin problems and postdelivery treatments. The plant juice is taken as an anti-
diabetic drug [13]. It is good against toothache too. The essential oil from S. crispus
can be used as a nutraceutical towards degenerative diseases [14]. The high content
of calcium carbonate in the plant makes the boiled water mildly alkaline and func-
tion in ease of urination [15]. A survey in Malaysia revealed its application to treat
kidney stones by placing heated leaves on hips [16]. Traditional medicine in
Malaysia and Indonesia uses filtrates of boiled Strobilanthes leaves as antidiabetic,
diuretic, antilytic, and laxative [2]. Blood sugar, leprosy, urinal problems, jaundice,
inflammation, and excess menstruation can be treated using S. heyneanus roots and
leaves [17].
The genus, Strobilanthes, has been widely used in traditional herbal practices of
India, China, Indonesia, etc. Moreover, its leaf extract can minimize the glucose
level in blood and also reduces the risk of heart muscle/cardiovascular ailment [18].
Empirical shreds of evidences support the use of traditional herbs that cured the
disease of thousands, but the role of many of these herbs remains largely lacking.
Strobilanthes: A Plethora of Phytomedicine 547
3 Phytochemical Constituents
chavicol. However, the oil from pre-flowering plants contains all four compounds
except methyl chavicol [26].
Anticestodal drugs are used mainly against tapeworms. With the help of
Hymenolepis diminuta (rat tapeworm) experimental model, anticestodal efficacy of
S. discolor leaf extract was investigated. It was analyzed by checking the eggs per
gram of feces counts and percentage worm recovery rates. It was followed by treat-
ing different groups of rats infected by Hymenolepis diminuta with methanol leaf
extract. There was a significant reduction in both parameters considered. Interesting
results were obtained in the larval stages of the parasite. In this stage, not a single
worm had escaped from the 800 mg/kg dose, which is given twice daily for a con-
tinuous three days. These findings suggest the high potential of the plant against
cestodes and support its use in folk medicine [27].
The GC-TOF mass spectroscopy of the methanolic and aqueous extracts of
S. crispus dried leaves of the plant revealed potential phytoconstituents [28]. About
32 compounds were identified such as 3-octadedecene, α-sitosterol, campesterol,
hexadecanoic acid methyl ester, lupeol, phytol, and stigmasterol in the methanolic
extract, while in the aqueous extract, 3,5-dithiahexanol 5,5-dioxide, cyclobutanol,
hydrazine carboxamide, monoethanolamine, n-propyl acetate, and undecane have
been identified. Moreover, the plant leaf contains ester glycosidic compound of caf-
feic acid (a verbascoside), voumaric acid, caffeic acid, vanilic acid, ferulic acid, and
syringic acids [19].
The wound healing capacity of S. crispus was studied in the rat. The wounds
dressed with leaf extracts showed less inflammatory cells and high collagen in his-
tological analysis [29]. The antiviral activity of human coronavirus
NL63(HCoV-NL63) using extracts of S. crispus leaf and its major components
revealed a positive result. Among the components, trypanthrin and indigodole B (5a
R-ethyl trypanthrine) were potential against viral activity and thereby reduce the
cytopathic effect and progeny virus production [30]. The HPLC analysis of S. cris-
pus opens up eight flavonoid compounds. They were catechin, epicatechin, rutin,
myricetin, luteolin, apigenin, naringenin, and kaempferol [31]. The compounds
from S. crispus will cohere the proteins that bind to the active part of the reverse
transcriptase. It inhibits retrovirus proliferation which causes acquired immune
deficiency syndrome (AIDS) and Adult T-cell leukemia [32]. The isolated and iden-
tified compounds in the taxa are summarized (Table 1).
The synergistic activity of alkaloids, steroids, terpenoids, phenols, flavonoids,
tannins, saponins, glycosides, and carbohydrates are together responsible for the
antioxidant and antibacterial efficacy of S. heyneanus [17]. A significant amount of
phenols, flavonoids, and tannins were observed in aqueous plant extract rather than
ethanol extract [33]. In S. cilliatus, triterpenoids were present in petroleum ether
and benzene-soluble parts, flavonoids and phenols in water, and alcohol-soluble
parts, whereas mucilage was present in water-soluble parts only [34].
Strobilanthes: A Plethora of Phytomedicine 549
4 Bioactivities
4.1 Antioxidant Activity
Antioxidants are the only way to fight against the double-edged sword ‘reactive oxy-
gen species’. The wise use of reduction of them can delimit the diseases to an extent.
Plant-derived secondary metabolites such as flavonoids, phenolic acids, and tannins
are well-known scavenging activators against reactive species. The oxidative human
diseases such as cancer, tissue damage, and DNA injury can be controlled with the
help of antioxidants. The ability of phytocompounds against these necessary evils is
well researched and discussed from time immemorial. Any substance which is pres-
ent in low concentrations compared to an oxidizable substrate and delays or inhibits
the oxidation of that substrate is called a biological antioxidant. An ideal antioxidant
at the physiological level will be completely absorbed by the body and undertake
quenching of free radicals or chelate redox metals [35] (Fig. 1).
Reactive oxygen species (ROS) are of both free radical and non-free radical
oxygen-containing species, which increase oxidative stress and impair the redox
balance [36]. They include hydrogen peroxide (H2O2), superoxide (O2-), singlet
oxygen (½O2), hydroxyl radical (∙OH), and reactive nitrogen, iron, copper, and sul-
phur species. Among them, hydroxyl radical is a potent ROS that causes cell dam-
age by reacting with phospholipids of the cell membrane. Superoxide is a weak one
that produces hazardous hydroxyl radicals and singlet oxygen [37]. According to
Sies [38], the oxidative stress is a disturbance in the prooxidant to antioxidant bal-
ance in service of the former, leading to potential damage. The oxidative stress
experienced by a cell depends on the activity of ROS generated and the ROS scav-
enging system. For favoring mild oxidative stress, the balance between prooxidant
and antioxidant substances is kept slightly in favor of prooxidant products [36].
The consumption of leaf extract of S. crispus as herbal tea increased the defense
system especially towards degenerative diseases due to the high number of water-
soluble vitamins [20]. The wide spectrum of its activities includes antiviral, antitu-
mor, anti-inflammatory, and anticoagulant [39]. The plant has been suggested as a
potential resource of squalene [40]. The aqueous extracts of S. crispus leaves col-
lected from North-east Malaysia (Kelantan) showed 73.8% in DPPH radical scav-
enging assay and 267.5 μM of Fe (II) activity in ferric reducing antioxidant power
assay (FRAP) with an IC50 value of 44.1 μg/mL [3].
The highest percentage of inhibition in antioxidant property of S. kunthianus
ethanolic extract (79.23 ± 0.37) was 250 μg/mL and followed by methanolic extract
(90.35 ± 0.54) against ascorbic acid as standard [41]. They suggested it as an herbal
alternative for various diseases. In S. barbatus, the concentration of the extract for
fifty percentage inhibition for DPPH, superoxide, and hydroxyl radical scavenging
activity were 15, 250, and 525 μg/mL, respectively [42].
The reduced awareness of pain is called analgesia. The analgesic effect of etha-
nolic extracts of S. cilliatus against the standard pain killer diclofenac is proved by
the tail clip method in Swiss Albino mice [43]. A strong correlation between anti-
oxidant activity and the total phenol contents was proposed by Qader et al. in 2011
[44]. S. crispus extracts showed nontoxic effects against a normal human lung fibro-
blast cell line (Hs888Lu). The ethanolic extracts contain high antioxidant activities
compared to aqueous extracts in 1,1-diphenyl-1-picrylhydrazyl (DPPH) and FRAP
assays. The methanolic extracts of S. crispus offer a richer source of dietary antioxi-
dants [45]. The convective dried S. crispus showed potential for preserving antioxi-
dant constituents by achieving the highest antioxidant values that were significantly
higher than those obtained after freeze-drying [46].
The antioxidant properties of aqueous and methanolic extracts were studied
using DPPH free radical, xanthine oxidase activity, and β-carotene-linoleate model
system in S. crispus by Muslim et al. [28]. The inhibitory activity of the S. crispus
extracts towards the xanthine oxidase enzyme was high but they demonstrated mod-
erate antioxidant properties. It can be witnessed by the quenching of the DPPH-free
radical and preventing the bleaching of β-carotene by linoleic acid. The higher anti-
oxidant activity of methanolic extract of S. glutinosus can be correlated to the high
content of phenol (247.85 mg GAE/g extract) and flavonoid (71.91 mg QAE/g
552 R. Chembrammal et al.
extract). But the n-hexane extract exhibited the highest anti-urease activity with IC50
value of 0.244 mg/ml [47] (Fig. 2).
In the case of S. kunthiana flowers, the ethyl acetate extract and n-butanol extract
exhibited promising antioxidant activity and the n-hexane extract was devoid of any
activity [48]. In the ABTS method, the ethyl acetate extracts of the root and stem of
the plant showed potent in vitro antioxidant activity. Among the different extracts
studied, crude methanolic flower extract was the most potent one. The weak cyto-
toxic activity against Hep2 and HeLa cell lines was exhibited by all the extracts
studied [49]. A dose-dependent activity was shown by ethanolic extract of S. asper-
rimus in serum biochemical parameters like hepatic antioxidant enzymes like gluta-
thione peroxidase (GPX), superoxide dismutase (SOD), catalase (CAT), and
malondialdehyde (MDA). When compared with the standard silymarin, significant
antioxidant activity was detected by decreasing MDA and increasing SOD, GPX,
and CAT [50]. The significant increase in glutathione peroxidase and superoxide
dismutase activities proves the antioxidant effect in streptozotocin-induced diabetic
rats treated with S. crispus juice [51].
4.2 Antidiabetic Activity
4.3 Antimicrobial Activity
4.4 Antiproliferative Activity
S. callosus has protective efficacy towards acute and chronic inflammation in rat
models [85]. They administered ethanol, chloroform, and petroleum ether extracts
in different doses (100, 200, and 400 mg/kg). The inflammation was induced by
carrageenan and Freund’s complete adjuvant model in the plantar surface of rats. In
the carrageenan-induced model, petroleum ether extract showed a significant effect,
whereas in Freund’s complete adjuvant model, both extracts were found to be effec-
tive. In vivo protection of CRC of S. crispus extract was done in HT29 cell lines [86].
According to Fernández et al. [87], pentacyclic terpenes could decrease edema
formation. Roots of S. callosus and S. ixiocephala have long been used for inflamma-
tion disorders in folk medicine [68] and their medicinal property is thought to be
conferred by pentacyclic terpene, lupeol. Lupeol at its effective doses exhibits no
toxicity to normal cells Vietnam [88]. Baraya et al. [89] studied the anti-migration,
anti-invasion, and anti-metastasis effects of S. crispus leaves on breast cancer cells
(MDA-MB-231) by using a subfraction F3. The fraction contains β-sitosterol, stig-
masterol, campesterol, lutein, pheophytin a, 131-hydroxy-132-oxo-pheophytin a, and
132-hydroxy-pheophytin a. Thus, the fraction exerted an antiproliferative effect with
the IC50 value of 84.27 μg/mL after 24 h of exposure and 74.41 μg/mL after 48 h of
exposure. Similarly, the subfraction F3 from S. crispus was capable of triggering the
immune system in rats-bearing NMU-induced mammary tumor. So, this may support
the traditional use of leaves of the plant to boost the immune system [90]. Another
study conducted by Gordani et al. [91] investigated the antiproliferative effects of
S. crispus on MCF-7 cells using different extracts of leaves and the stem (methanol,
hexane, chloroform, ethyl acetate, and aqueous extracts). The essential oil from
S. crispus did not show any effect on both MCF-7 and MDA-MB-231 cells [74].
The dichloromethane fraction (F3) of S. crispus was evaluated for the immuno-
modulatory effects. The administration of F3 enhanced the expression of CIITA and
MHC-II on the mammary cancer cells and the number of infiltrating CD4+ and
CD8+ immune cells. Along with that after 2 months, the serum level of chemokine
ligand 2 (CCL2) decreased significantly, while the level of interferon-gamma (IFN-
γ) increased [90]. Among the five different leaf extracts of S. crispus studied (hex-
ane, chloroform, ethyl acetate, methanol, and aqueous) in CNE-1 cells, the ethyl
acetate extract showed the strongest antiproliferative effect on the cells with an
IC50 value of 119.00 ± 48.10 μg/mL. It was followed by an IC50 value of
119.00 ± 48.10 μg/mL [92]. The previous cell line studies conducted in the taxa are
tabulated (Table 2).
There are two types of lung cancers: non-small cell lung cancer and small cell
lung cancer. The symptoms include a slight cough or shortness of breath and may
become severe. The methanolic leaf extract of S. crispus was analyzed for its cyto-
toxic effect on the NCI-H23 lung cell line. But there was no considerable effect in
it with an IC50 value greater than 200 μg/mL [93]. The methanolic extract of S. cris-
pus exhibited a cytotoxic response towards the T-47D and MCF7 cells and the aque-
ous extract was found to be nontoxic towards all cell lines used. A notable
antiangiogenic activity was shown by both aqueous and methanolic extracts [28].
There was a significant decrease in serum glucose levels in both male and female
diabetic and normal rats when treated with S. crispus juice. It has reduced the
Strobilanthes: A Plethora of Phytomedicine 557
5 Conclusion
Phytotherapy or herbal medicine or herbalism is the use of plants’ extracts for either
treatment or health-promoting purposes. The phytochemical constituents synergi-
cally act each other to enhance the bioactivity and pharmacological efficacy of a
plant. Most of the Strobilanthes species are underexplored. The various potential
bioactivities of the plant support their use in traditional medicine. Despite the medic-
inal properties, the economic utility of the genus is not explored. The present study
highlights the pharmacological value of the Strobilanthes species in the literature.
Thus, it opens up a wide area of future research in the taxa in drug discoveries.
Acknowledgments The first author acknowledges the financial grant supported by the Council of
Scientific and Industrial Research (CSIR) in the form of Senior Research Fellowship
(09/043(0186)/2017-EMR-1). The second author acknowledges University of Calicut for the
financial assistance rendered.
Declarations The authors declare no conflict of interest.
558 R. Chembrammal et al.
References
1. Sivarajan V, Balachandran I (1994) Ayurvedic drugs and their plant sources. Oxford and IBH
Publishing Co. Pvt. Ltd, New Delhi
2. Bakar MFA, Teh AH, Rahmat A, Hashim N, Othman F, Fakurazi S (2006) Antiproliferative
properties and antioxidant activity of various types of Strobilanthes crispus tea. Int J Cancer
Res 2:152–158
3. Ghasemzadeh A, Jaafar HZ, Rahmat A (2015) Phytochemical constituents and biological
activities of different extracts of Strobilanthes crispus (L.) Bremek leaves grown in different
locations of Malaysia. BMC Complement Altern Med 15(1):1–10
4. Li XB, Komatsu K, Zhou GC, Namba T (1997) Pharmacological studies on the Chinese
crude-drug “Shengma” (IV) 1. Nat Med 51:408–416
5. Chen H, Shao J, Zhang H, Jiang M, Huang L, Zhang Z et al (2018) Sequencing and analy-
sis of Strobilanthes cusia (Nees) Kuntze chloroplast genome revealed the rare simultaneous
contraction and expansion of the inverted repeat region in angiosperm. Front Plant Sci 9:324.
https://doi.org/10.3389/fpls.2018.00324
6. Xu W, Zhang L, Cunningham AB, Li S, Zhuang H, Wang Y, Liu A (2020) Blue genome:
chromosome-scale genome reveals the evolutionary and molecular basis of indigo biosynthe-
sis in Strobilanthes cusia. Plant J 104(4):864–879
7. Wu W, Li J, Liu Y, Jiang M, Lan M, Liu C (2021) Peculiarities of the inverted repeats in the
complete chloroplast genome of Strobilanthes bantonensis Lindau. Mitochondrial DNA Part
B 6(4):1440–1447
8. Chiang YR, Li A, Leu YL, Fang JY, Lin YK (2013) An in vitro study of the antimicrobial
effects of indigo naturalis prepared from Strobilanthes formosanus Moore. Molecules
18:14381–14396
9. Jain SK, Defilipps RA (1991) Medicinal plants of India (pp. 92). Reference Publications,
Inc., Algonac Michigan
10. Warrier PK, Nambiar VPK, Raman Kutty C (1994) Indian medicinal plants, vol 5. Orient
Longman Ltd, Madras, pp 142–145
11. Thomas J, Joy PP, Mathew S, Skaria BP, Duethi PP, Joseph TS (2000) Agronomic practices
for aromatic and medicinal plants. Kerala Agricultural University, Calicut
12. Fatima I, Ahmad I, Anis I, Malik A, Afza N (2007) Isatinones a and B, new antifungal oxin-
dole alkaloids from Isatis costata. Molecules 12:155–162
13. George M, Joseph L, Sony S (2017) A review on pharmacological and biological activities of
Strobilanthes ciliatus Nees. World J Pharm Pharm Sci 6:504–512
14. Rahmat A, Edrini S, Ismail P, Taufiq Y, Yun H, Abu Bakar MF (2006) Chemical constitu-
ents, antioxidant activity and cytotoxic effects of essential oil from Strobilanthes crispus and
Lawsonia inermis. J Biol Sci 6:1005–1010
15. Noraida A (2005) Penyembuhan semula jadi dengan herba. Medicinal Plants Malaysia. Herbs
Thearapic Use in Malaysia
16. Ong HC, Norzalina J (1999) Malay herbal medicine in Gemencheh, Negeri Sembilan.
Malaysia Fitoterapia 70(1):10–14
17. Sundaram V, Sadhasivam S, Chandrasekaran S, Nanjian R, Pandian A (2021) Strobilanthes
heyneanus root extract as a potential source for antioxidant and antimicrobial activity. Future
J Pharm Sci 7(1):1–12
18. Fadzelly AM, Asmah R, Fauziah O (2006) Effects of Strobilanthes crispus tea aqueous
extracts on glucose and lipid profile in normal and streptozotocin-induced hyperglycemic
rats. Plant Foods Hum Nutr 61(1):6–11
19. Cheong BE, Zakaria NA, Cheng AYF, Teoh PL (2016) GC-MS analysis of Strobilanthes
crispus plants and callus. Trans Sci Technol 3(1–2):155–161
20. Maznah I, Manickam E, Danial AM, Rahmat A, Yahaya A (2000) Chemical composition and
antioxidant activity of Strobilanthes crispus leaf extract. J Nutr Biochem 11:536–542. https://
doi.org/10.1016/S0955-2863(00)00108-X
Strobilanthes: A Plethora of Phytomedicine 559
21. Lee CL, Wang CM, Hu HC, Yen HR, Song YC, Yu SJ et al (2019) Indole alkaloids ind-
igodoles A-C from aerial parts of Strobilanthes cusia in the traditional Chinese medicine
Qing Dai have anti-IL-17 properties. Phytochemistry 162:39–46. https://doi.org/10.1016/j.
phytochem.2019.02.016
22. Gu W, Wang W, Li XN, Zhang Y, Wang LP, Yuan CM, Huang LJ, Hao XJ (2015) A novel iso-
coumarin with anti-influenza virus activity from Strobilanthes cusia. Fitoterapia 107:60–62.
https://doi.org/10.1016/j.fitote.2015.10.009
23. Laitonjam WS, Wangkheirakpam SD (2011) Comparative study of the major components
of the indigo dye obtained from Strobilanthes flaccidifolius Nees. And Indigofera tinctoria
Linn. Int. J Plant Physiol Biochem 3(5):108–116. https://doi.org/10.5897/IJPPB.9000010
24. Lim KT, Lim V, Chin JH (2012) Subacute oral toxicity study of ethanolic leaves extracts
of Strobilanthes crispus in rats. Asian Pac J Trop Biomed 2(12):948–952. https://doi.
org/10.1016/S2221-1691(13)60005-2
25. Fernandes MC, Krishnan S (2019) GC-MS analysis of metabolites in leaf and stem of medic-
inally important Strobilanthes species from North Western Ghats of India. Phytomorphology
69:67–79
26. Weyerstahl P, Marschall-Weyerstahl H, Manteuffel E, Kaul VK (1992) Constituents of the
essential oil of Strobilanthes callosus Nees. J Essent Oil Res 4(3):281–285. https://doi.org/1
0.1080/10412905.1992.9698062
27. Tangpu V, Yadav AK (2006) Anticestodal property of Strobilanthes discolor: an experimental
study in Hymenolepis diminuta - rat model. J Ethnopharmacol 105(3):459–463. https://doi.
org/10.1016/j.jep.2005.11.015
28. Muslim NS, Ng KW, Itam A, Nassar ZD, Ismail Z, Abdul Majid AMS (2010) Evaluation of
cytotoxic, anti-angiogenic and antioxidant properties of standardized extracts of Strobilanthes
crispus leaves. Int J Pharmacol 6(5):591–599
29. Al-Henhena N, Mahmood AA, Al-Magrami A, Syuhada AN, Zahra AA, Summaya MD, Suzil
MS, Salmah I (2011) Histological study of wound healing potential by ethanol leaf extract
of Strobilanthes crispus in rats. J Med Plant Res 5(16):3660–3666. https://doi.org/10.5897/
JMPR.9000704
30. Tsai YC, Lee CL, Yen HR, Chang YS, Lin YP, Huang SH, Lin CW (2020) Antiviral action of
tryptanthrin isolated from Strobilanthes cusia leaf against human coronavirus NL63. Biomol
Ther 10(3):366. https://doi.org/10.3390/biom10030366
31. Liza MS, Rahman RA, Mandana B, Jinap S, Rahmat A, Zaidul ISM, Hamid A (2010)
Supercritical carbon dioxide extraction of bioactive flavonoid from Strobilanthes cris-
pus (Pecah Kaca). Food Bioprod Process 88(2–3):319–326. https://doi.org/10.1016/j.
fbp.2009.02.001
32. Endrini S, Rahmat A, Ismail P, Taufiq-Yap YH (2007) Comparing of the cytotoxicity proper-
ties and mechanism of Lawsonia inermis and Strobilanthes crispus extract against several
cancer cell lines. J Med Sci 7(7):1098–1102
33. Kavitha CCI, Indira G (2016) Quantitative estimation of total phenolic, flavonoids, tannin and
chlorophyll content of leaves of Strobilanthes kunthiana (Neelakurinji). J Med Sci 4:282–286
34. Shirwaikar A, Sajith Kumar PN, Rajagopal PL (2018) Preliminary phytochemical and
physicochemical analysis on Strobilanthes ciliatus Nees. Research review. Int J Multidiscip
3(4):125–127
35. Halliwell B, Gutteridge JMC (1999) Free radicals in biology and medicine. Clarendon
Press, Oxford
36. Poljsak B, Šuput D, Milisav I (2013) Achieving the balance between ROS and antioxidants:
when to use the synthetic antioxidants. Oxidative Med Cell Longev 2013:1. https://doi.
org/10.1155/2013/956792
37. Alam MN, Bristi NJ, Rafiquzzaman M (2013) Review on in vivo and in vitro methods evalu-
ation of antioxidant activity. Saudi Pharma J 21(2):143–152
38. Sies H (1991) Oxidative stress: from basic research to clinical application. Am J Med
91(3):31–38
560 R. Chembrammal et al.
39. Chen TQ, Wu YB, Wu JG, Wang HY, Mao FH, Wu JZ (2013) Fatty acids, essential oils,
and squalene in the spore lipids of Ganoderma lucidum by GC-MS and GC-FID. Chem Nat
Compd 49:143. https://doi.org/10.1007/s10600-013-0536-x
40. Yang T, Wu Q, Li SY, Lv ZJ, Hu B, Xie BJ, Sun ZD (2014) Liposoluble compounds with anti-
oxidant activity from Strobilanthes tonkinensis. Chem Nat Compd 49(6):1166–1167. https://
doi.org/10.1007/s10600-014-0852-9
41. Prabakaran R, Kirutheka E (2018) GC-MS, phytochemicals and antioxidant activities of in
vitro callus extracts of Strobilanthes kunthiana (Nees) T. Anderson ex Benth: an endemic
plant of Acanthaceae. Braz. J Biol Sci 5(10):359–372
42. Subbulekshmi KO, Godwin SE, Vahab AA (2015) Phytochemical and in-vitro antioxidant
activity of ethanolic extract of Strobilanthes barbatus Nees leaves. Asian J Pharm Res Dev
3(2):13–20
43. Mathew G, Lincy J, Sony S (2017) Evaluation of analgesic activity of ethanolic extract of
Strobilanthes ciliatus Nees. Pharma Innov 6(7):326–328
44. Qader SW, Abdulla MA, Chua LS, Najim N, Zain MM, Hamdan S (2011) Antioxidant,
total phenolic content and cytotoxicity evaluation of selected Malaysian plants. Molecules
16(4):3433–3443. https://doi.org/10.3390/molecules16043433
45. Tan HM, Leong KH, Song J, Mohd Sufian NSF, Mohd Hazli UHA, Chew LY, Kong KW
(2020) Antioxidant and LC-QToF-MS/MS analysis of polyphenols in polar and non-polar
extracts from Strobilanthes crispus and Clinacanthus nutans. Int Food Res J 27(5):903–914
46. Chua LYW, Chua BL, Figiel A, Chong CH, Wojdyło A, Szumny A, Choong TSY (2019)
Antioxidant activity, and volatile and phytosterol contents of Strobilanthes crispus dehy-
drated using conventional and vacuum microwave drying methods. Molecules 24. https://doi.
org/10.3390/molecules24071397
47. Aziz M, Ahmad S, Iqbal MN, Khurshid U, Saleem H, Rehman KU, Alamri A, Anwar S,
Alamri AS, Chohan TA (2021) Phytochemical, pharmacological, and in-silico molecular
docking studies of Strobilanthes glutinosus Nees: an unexplored source of bioactive com-
pounds. S Afr J Bot. https://doi.org/10.1016/j.sajb.2021.07.013
48. Singh BB, Das S, Maithi A (2014) Antioxidant property for lipophilic extract of Strobilanthes
kunthiana flowers. Indian J Res Pharm Biotechnol 2(1):1005–1009
49. Balasubramaniam G, Sekar M, Badami S (2021) In-vitro antioxidant and cytotoxic properties
of Strobilanthes kunthianus. Res J Pharm Technol 14(5):2522–2528
50. Samal PK (2013) Antioxidant activity of Strobilanthes asperrimus in albino rats. Asian J
Pharm Clin Res 3:71–74
51. Norfarizan-Hanoon NA, Asmah R, Rokiah MY, Fauziah O, Faridah H (2009)
Antihyperglycemic, hypolipidemic and antioxidant enzymes effect of Strobilanthes crispus
juice in normal and streptozotocin-induced diabetic male and female rats. Int J Pharmacol
5(3):200–207
52. Maritim AC, Sanders A, Watkins JB III (2003) Diabetes, oxidative stress, and antioxidants: a
review. J Biochem Mol Toxicol 17(1):24–38. https://doi.org/10.1002/jbt.10058
53. Mukherjee PK, Maiti K, Mukherjee K, Houghton PJ (2006) Leads from Indian medici-
nal plants with hypoglycemic potentials. J Ethnopharmacol 106(1):1–28. https://doi.
org/10.1016/j.jep.2006.03.021
54. Samal PK (2013) Antidiabetic and antioxidant activity of strobilanthes asperrimus in rats. J
Glob Trends Pharma Sci 4(2):1067–1072
55. Joseph S, Kumar L, Bai VN (2016) Evaluation of anti-diabetic activity of Strobilanthes cus-
pidata in alloxan induced diabetic rats and the effect of bioactive compounds on inhibition of
α-amylase enzyme. J Pharmacogn Phytochem 5(3):169–175
56. Balouiri M, Sadiki M, Ibnsouda SK (2016) Methods for in vitro evaluating antimicrobial
activity: a review. J Pharm Anal 6(2):71–79. https://doi.org/10.1016/j.jpha.2015.11.005
57. Lim V, Yap CS, Chong HW, Shukkoor MSA, Priya M (2013) Antimicrobial evaluation and
GC-MS analysis of Strobilanthes crispus ethanolic leaf extract. Methodology 10(4):1–8.
https://doi.org/10.9734/EJMP/2015/20075
Strobilanthes: A Plethora of Phytomedicine 561
76. Yaacob NS, Hamzah N, Kamal NNNM, Abidin SAZ, Lai CS, Navaratnam V, Norazmi MN
(2010) Anticancer activity of a sub-fraction of dichloromethane extract of Strobilanthes
crispus on human breast and prostate cancer cells in vitro. BMC Complement Altern Med
10(1):1–14. https://doi.org/10.1186/1472-6882-10-42
77. Strobel G, Daisy B (2003) Bioprospecting for microbial endophytes and their natural products.
Microbiol Mol Biol Rev 67:491–502. https://doi.org/10.1128/MMBR.67.4.491-502.2003
78. Jinfeng EC, Rafi MIM, Hoon KC, Lian HK, Kqueen CY (2017) Analysis of chemical constit-
uents, antimicrobial and anticancer activities of dichloromethane extracts of Sordariomycetes
sp. endophytic fungi isolated from Strobilanthes crispus. World J Microbiol Biotechnol
33(1):1–19. https://doi.org/10.1007/s11274-016-2175-4
79. Muhammad SN, Yaacob NS, Safuwan NA, Fauzi AN (2021) Antiglycolytic activities
of Strobilanthes crispus active fraction and its bioactive components on triple-negative
breast cancer cells in vitro. Anti Cancer Agents Med Chem 22:1363. https://doi.org/10.217
4/1871520621666210427104804
80. Endrini S, Rahmat A, Ismail P, Taufiq-Yap YH (2014) Cytotoxic effect of γ-sitosterol from
Kejibeling (Strobilanthes crispus) and its mechanism of action towards c-myc gene expres-
sion and apoptotic pathway. Med J Indones 23(4):203–208. https://doi.org/10.13181/mji.
v23i4.1085
81. Paulsamy S, Vijayakumar KK, Murugesan M, Padmavathy S, Senthilkumar P (2007)
Ecological status of medicinal and other economically important plants in the shola under-
stories of Nilgiris, the Western Ghats. Nat Prod Radiance 6:55–61
82. Desu BSR, Elango K, Satish Kumar MN, Suresh B, Manimaran S, Nanjan MJ (2011)
In-vitro anti-inflammatory and anti-osteoarthritic activities of Strobilanthes kunthianus and
Strobilanthes cuspidatus. Int J Res Pharm Chem 1(4):1265–1268
83. Desu BS, Elango K, Satish Kumar MN, Suresh B, Manimaran S, Nanjan MJ (2011)
Investigation of selected medicinal plants (Strobilanthes kunthianus, Strobilanthes cuspida-
tus) and marketed formulation (Shallaki) for their anti-inflammatory and anti-osteoarthritic
activity. Pharmanest 2:492–499
84. Balasubramaniam G, Sekar M, Varadarajan M, Badami S (2020) Antioxidant and hepatopro-
tective activities of Strobilanthes kunthianus against carbon tetrachloride-induced hepatotox-
icity in rats. Pharm J 12(5):1143–1151. https://doi.org/10.5530/pj.2020.12.161
85. Kumar S, Bhatia M, Garg LN, Gupta S (2013) Acute and chronic inflammation studies of
Strobilanthes callosus leaves extract on rat model. Inflammopharmacology 21(3):233–239.
https://doi.org/10.1007/s10787-012-0150-8
86. Al-Henhena N, Khalifa SA, Ying RPY, Ismail S, Hamadi R, Shawter AN, Idris AM, Azizan A,
Al-Wajeeh NS, Abdulla MA, El-Seedi HR (2015) Evaluation of chemopreventive potential of
Strobilanthes crispus against colon cancer formation in vitro and in vivo. BMC Complement
Altern Med 15:419. https://doi.org/10.1186/s12906-015-0926-7
87. Fernández MA, de las Heras B, Garcia MD, Sáenz MT, Villar A (2001) New insights into
the mechanism of action of the anti-inflammatory triterpene lupeol. J Pharm Pharmacol
53(11):1533–1539
88. Siddique HR, Saleem M (2011) Beneficial health effects of lupeol triterpene: a review of
preclinical studies. Life Sci 88(7–8):285–293. https://doi.org/10.1016/j.lfs.2010.11.020
89. Baraya YS, Wong KK, Yaacob NS (2019) Strobilanthes crispus inhibits migration, invasion
and metastasis in breast cancer. J Ethnopharmacol 233:13–21. https://doi.org/10.1016/j.
jep.2018.12.041
90. Yankuzo HM, Mustapha Z, Wong KK, Yaacob NS (2018) Immunomodulatory effects of a
bioactive fraction of Strobilanthes crispus in NMU-induced rat mammary tumor model. J
Ethnopharmacol 213:31–37. https://doi.org/10.1016/j.jep.2017.10.024
91. Gordani N, Cheong BE, Teoh PL (2017) Antiproliferative effect of Strobilanthes crispus on
MCF-7 cell line. Trans Sci Technol 4:414–419
92. Koh RY, Sim YC, Toh HJ, Liam LK, Ong RSL, Yew MY, Tiong YL, Ling APK, Chye SM,
Ng KY (2015) Cytotoxic and apoptogenic effects of Strobilanthes crispa Blume extracts
Strobilanthes: A Plethora of Phytomedicine 563
1 Introduction
Cancer continues to claim lives globally, and as per data published by WHO, in
2020, cancer claimed ten million lives that accounts for one in every six deaths and
it is the second leading cause of death in the world. Cancer affects human around
the globe with no bars to color, creed, or geographical barriers. Though the number
of people receiving a cancer diagnosis has steeply increased over the years, the sur-
vival rates are improving remarkably. The scientists and research scholars in this
field are dedicated to develop various noninvasive drugs for treating cancer. The
mental and physical agony caused due to the side effects of conventional treatment
methods like chemotherapy and anticancer drugs are beyond words to describe and
continue to haunt patients even after recovery form the disease. During treatment,
patients suffer from anemia, appetite loss, hair loss (Alopecia), nerve problems
(Peripheral neuropathy), organ-related inflammation, sleep problems, and insom-
nia. In this context anticancer drugs of natural origin, herbal remedies, and natural
phytochemicals have been the focus area for managing the adverse side effects of
cancer treatment. Plant-based anticancer drugs containing bioactive compounds of
medicinal plants like vinblastine, taxol, vincristine, epipodophyllotoxins, and camp-
tothecine derivatives have proved efficacy in treating human cancers. Most interest-
ingly, some of these drugs act like a natural defensive mechanism against formation
of cancer cells in human body. This throws light into the arena of cancer prevention
rather than cancer treatment with phytochemicals derived from medicinal plants
which we see around us.
Annona cherimola is an erect plant with long and light green leaves and white,
fleshy fragrant flowers. Fruits are conical, oval, or heart-shaped with fingerprint-like
markings on its surface. Snow-white juicy flesh contains oval-oblong, slightly flat-
tened brown or black, bean-like, glossy seeds. Annona crassiflora is a deciduous
tree having ovate leaves, their yellow-greenish leathery flowers have a jellyfish like
appearance. Fruits are sweet and have fibrous, succulent, and creamy pulp. Annona
dioica is a small shrub with oval, light green hairy leaves. Fleshy, leathery leaves
and yellowish flowers are present. Yellowish green spiky edible fruits are present in
the plant with a creamy pulp inside it. Annona glabra is semi-deciduous tropical
plant having ovate to oblong leaves in it. Leaves are glossy and hairless having the
smell similar to green apples. Annona montana is an evergreen or semievergreen
shrub or tree with large glossy and glabrous leaves and slightly scented flowers. Its
fruits are dark-green in color, studded with spines, and have yellow-orange flesh. Its
seeds are light-brown and waxy in nature. A. glabra have creamy or pale-yellow
leathery flowers. The mango-shaped fruits having smooth and shiny surface are
edible and it contains pinkish-orange pulp with many light yellows to brown seeds.
Annona muricata is small, evergreen plant with green glossy leaves which are
oblong to oval and slightly hairy branches. Flowers are thick and yellowish which
appear singly on a woody stalk on twigs, branches, or trunks. Fruits contain black
colored seeds in a creamy white flesh inside it with cotton-like fibers. They are
heart-shaped with curved spines covering the leathery rough skin and dark green in
color. Annona reticulata is a small shrub or semievergreen tree. Leaves are alternate
and hairless pointed at the apex. Flowers are yellow-green in color generally seen in
clusters. The reddish-yellow to brown fruitsvary in shape, heart-shaped, spherical,
or oblong having white pulp. Blackish brown glossy seeds are present in the fruit
and it is covered in polygonal plates. Annona purpurea is a deciduous shrub or tree
with hairy, elliptical, or oval-shaped leaves and large, strong-scented flowers. The
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 567
edible fruit is rounded with a felt-textured brown skin and having hook like projec-
tions on the surface. The fruit of the plant appears as stringy and fibrous. Bigger
spiky and hard-shelled fruits have yellowish orange sweet flesh with black seeds.
Annona squamosa is semi-deciduous shrub with very simple leaves having fine
hairs underneath and glaring greenish yellow pendulous flowers. The edible fruit is
greenish yellow in color with a knobby surface and have brown to black oblong
seeds embedded in soft creamy pulp. Annona senegalensis is a small tree with green
to bluish leaves without hairs on the top, but having brownish colored hairs under-
side. The flowers are yellowish in color, waxy, closely petalled, and very fragrant.
The fruits are yellow to orange in color, are sweet in taste, have pineapple-like odor,
and carry cylindrical orange-colored seeds.
Different Annonacea plant species can be seen in many parts of India and cap-
tures researcher’s attention due to its high nutritional value and medical applica-
tions. Pharmacological analysis on bark, fruit, leaves, and seeds of genus Annona
explored its anti-inflammatory, antitumor, anti-HIV, anticancer, antiprotozoal, anti-
diabetic, analgesics, antiparasitic, antihypertensive, gastroprotective, and hepato-
protective properties. Annonaceous acetogenins and alkaloids were potential
candidates believed to be responsible for significant biological activities [1].
Investigation on the discovery of anticancer agents from nature especially from this
plant origin is currently going on. Reports showed that annonaceous acetogenins
inhibits mitochondria complex I and NADH oxidase activity of plasma membranes
of tumor cells [2]. Selective suppression of cell growth of cancerous cells and drug
resistance cancerous cells in a dose-dependent manner were also reported [3].
Annonaceous acetogenins and alkaloids possessed antitumor potential and exhibit
cytotoxic effect through various mechanisms including inhibition of caspase, arrest-
ing the cell cycle, apoptosis, etc. [4]. Recent reports also showed acetogenins
extracted from the Annonacea family have pronounced cytotoxicity against respira-
tory complex [5]. Many studies have been reported till now elaborating the thera-
peutical applications of Annona genus as a potent anticancer agent. This chapter
comprises the reported antitumor activities of different Annonacea plant species
from Annona genus which showed potent activities in various types of cancer cells
in order to demonstrate their importance in cancer research and their mechanism of
action in detail [Fig. 1].
Annona cherimola commonly called cherimoya is a small deciduous tree with
edible fruits commonly found in the tropical or subtropical regions. Phytochemical
investigation of this plant has revealed the presence of flavonoids, alkaloids, glyco-
sides, tannins, steroids, saponins, amino acids, carbohydrates, proteins, and pheno-
lic compounds. Essential oil and ethanol extract of A. cherimola exhibited potent
anticancer effect against colon (CACO-2), liver (HEPG-2) and breast (MCF-7), and
carcinoma cell lines [6]. Two acetogenins namely, Aromin-A and squamocin, iso-
lated from methanolic extract of A. cherimola stem inhibited cell proliferation activ-
ity as reported in a study conducted in 1999 [7]. Seed extracts reported the
programmed cell death in Acute Myeloid Leukemia (AML) cell lines by the excita-
tion of both the intrinsic and extrinsic pathways [8]. Cytotoxic acetogenins, namely,
annomolin and annocherimolin reported from ethanolic extract of seeds of Annona
568 S. M. Joseph and A. R. Amala Dev
cherimola, was found to exhibit selectivity to human prostate tumor cell line (PC-3),
breast (MCF-7), and colon (HT-29) cancer cell lines, respectively [9, 10]. Previous
studies confirmed the potent inhibitory effect of terpene rich fraction of A. cheri-
mola leaf extract on Acute Myeloid Leukemia (AML), MDBK, and Hep-2cells
[11]. Evaluation of A. cherimola seed extract on human stomach gastric adenocar-
cinoma cell line [12], molvizarin, and motrilin which was the first C-29 hydroxyl-
ated acetogenin reported from the methanolic extracts of A. cherimola seeds
exhibited promising anticancer activity [13]. Bioactive acetogenins from A. cheri-
mola seeds proved its suppression efficiency similar to that of chemotherapeutic
drug namely Daunorubicin [14]. Cis-Potent cytotoxicity of annonacin, (2,4)-cis-and
trans-isoannonacins against pancreatic, prostate cancer cell line and almunequin,
cherimolin-2 reported from A. cherimola seeds against human KB carcinoma cell
line and monkey VERO cell line were reported [15, 16].
Another species Annona coriacea is an endemic species of the Brazilian Cerrado
that exhibits promising biological activities. A new acetogenin, coriadienin, is
reported from A. coriaea roots along with a known cytotoxic compound, gigantet-
ronenin. Coriadienin is attributed to the anticancer activity in VERO and KB cell
lines [17]. The extract showed autophagy inhibition in the cervical cancer cell line
which is an asset in cancer treatment. Acetogenins and alkaloids present in the
A. coriacea fraction were responsible for the cytotoxicity in cervical cancer cell
lines via DNA damage. Mode of action is found to be in a dose-dependent manner.
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 569
possessed very high anticancer activity against glioblastoma multiforme, lung car-
cinoma A-549, and breast adenocarcinoma [32]. Annoglabasin H, a component pre-
viously reported from A. glabra fruits, also showed significant cytotoxic activity in
SK-Mel2, LU-1, MCF-7, and KB with IC50 values ranging from 3.7 to 4.6 μM [33].
A. glabra extracts showed cytotoxicity to drug variant cancer cell lines. Seed extract
promoted cell arrest at the G0/G1 phase. Seed extract showed necrosis and apopto-
sis in both sensitive and resistant leukemia cells. The mode of action was found to
be in a concentration-dependent way [34]. A. glabra stem bark extract exhibited
cytotoxic activity and kaur-16-en-19-oic acid isolated by extract fractionation was
responsible for the therapeutical applications of the extract [35]. Previous studies
indicated that annomontacin being an anticancer compound caused variations in
mitochondrial transmembrane and induce apoptosis human liver cancer cell line
(Hep G2). The isolated diterpenoid compounds ent-kauran-19-al-17-oic acid
cunabic acid from A. glabra Linn was tested against liver Cancer (HLC) cell line
SMMC-7721 and found that it exhibited proliferation activity by apoptosis via
downregulating the gene expression of the BCL-2 gene and upregulating that of
Bax gene.
Annona hypoglauca, also known as wild cherimoya, were found in Amazon
floodplains. Chemical profile and bioactivity studies of A. hypoglauca are not much
available. Acid-base partitioning of A. hypoglauca stem yields aporphine alkaloids,
namely anonaine, actinodaphnine, isoboldine and nornuciferine. Actinodaphnine in
Annona species was reported for the first time and it was found that alkaloid-free
fraction does not show anticancer activity. Earlier reports indicated the cytotoxicity
of the crude extract against colon and breast cancer cells. Treatment of breast cancer
cell line by fraction containing isoboldine and actinodaphnine inhibited tumor for-
mation [36]. Cytotoxic potential of hexane and ethanolic extracts of leaves of
A. hypoglauca was investigated on Artemia salina and found that ethanolic extract
showed potent antitumor activity [37]. A plant Annona jahnii which is native to
Brazil, Colombia, and Venezuela is a tree having rich fruits. Acetogenins reported
from A. jahnii exhibited cytotoxic activities. Two new acetogenins, namely annodi-
enin and jahnonacin isolated from twigs of A. jahnii, indicated selective cytotoxicity
to six human tumor cell lines [38]. Another study showed that annojahnin isolated
from the twig found to exhibit potent cytotoxicity in six human solid cancerous cell
lines compared to standard drug Adriamycin [39].
Annona montana or mountain soursop was native to South America.
Annonacinand montanacin F isolated from leaves of A. montana showed in vivo and
in vitro cytotoxic activity, respectively, against the Lewis lung carcinoma (LLC) cell
lines [40]. Bioactive cytotoxic compounds such as iso-acetogenin, montanacin G,
three pairs of acetogenins, montanacin H-J and 34-epi-montanacin H-J, montana-
cins B and C, annonacin and cis-annonacin, gigantetrocins A and B were isolated
from A. montana leaves exhibited in vitro cytotoxic effect against Meth-A and LLC
tumor cell lines [41]. Further studies conducted in organic extracts of A. montana
fruit showed cytotoxicity on human astrocytoma, breast, lung, colon, and prostate
cancers [42]. Among the compounds montacin, cis-montacin, annonacin, cis-
annonacin, annomontacin and cis-annomontacin isolated from A. montana seeds,
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 571
effects against PACA-2 and HT-29 cell lines [72]. Muricin M and N from fruit pow-
der of A. muricata also possessed potent cytotoxicity against human prostate cancer
(PC-3) cells [73]. Cis annonacin from the seeds of A. muricata indicated selective
cytotoxicity to HT-29 cells with potency 10,000 times that of Adriamycin [74].
Antitumor potential of chloroform fraction of seeds was also confirmed [75].
Annona purpurea is commonly known as Soncoya, a common small to medium
deciduous tree best suited to tropical climates. New aporphine alkaloid
Oxoaporphine, isolated from A. purpurea, possessed moderate in vitro cytotoxicity
against 9-KB tumor [76]. Previous studies showed bis-THF acetogenins were the
most important components found in the roots of A. purpurea [77]. Annopurpuricin
A previously reported from A. purpurea dichloromethane extract of root indicated
mitochondrial necrosis, depolarization, and apoptosis in tumor cells. Purpuracenin,
annoglaucin purpurediolin, and purpurenin isolated from seeds of A. purpurea were
previously evaluated against various types of human cancer cells indicated in vitro
cytotoxic activity [78, 79]. Recently, a study quantified the anticancer agent, anno-
purpuricins, present in the plant using diode array high-performance liquid chroma-
tography detector (HPLC-DAD) [80]. Annona reticulata generally referred as
‘custard apple or bullock’s heart’ is a small, evergreen to the deciduous, tropical
tree. Phytoconstituents identified from the plant include flavanoida, alkaloids, tan-
nins, glycosides, steroids, amino acids, carbohydrates, acetogenins, etc. The plant
has several medicinal properties. Phytochemicals derived from different plant parts
are reported as potential anticancer compounds [81]. Evaluation of A. reticulata
extract indicated dose-dependent chemotherapeutic effect against HT-29, SCC9,
MCF-7, A549, and HCT116 [82–85]. Methanolic extract of leaves indicated cyto-
toxicity against breast carcinoma (T-47D), colon, hepatoma liver, and lung cancer
cell lines [86–90]. A study conducted in 2003 confirmed apoptotic cell death in T24
cells by annonacin isolated from the A. reticulata seeds [91]. Petroleum ether extract
of seeds was found to cause apoptosis in breast cancer cell line (MCF-7) and leuke-
mia cell line (K562) [92]. A previous study reported cytotoxic activity of ethanolic
extract of A. reticulata root against human lung, melanoma, chronic myelogenous
leukemia, cervix, and breast cancer cell line [93].
Annona salzmannii or Beach sugar apple is an extremely rare, tropical, evergreen
tree native to Brazil. Methanolic extract of A. salzmannii root yielded a new bis
tetrahydrofuran acetogenin, Salzmanin, along with previously known compounds
such as squamocin, almunequin, bullatalicin, and annonacin. Antitumor activities of
salzmanin and squamocin were reported [94]. A study conducted in 2013 showed
cytotoxic activity of essential oil isolated from A. salzmannii leaf extract [95].
Annona scleroderma, the poshe-te, is a species of tree in the Annonaceae family,
with an edible fruit native to the Atlantic coast of Central America, from Mexico
and Guatemala to Honduras. Methanol extract of A. scleroderma seeds yielded new
cytotoxic cyclononapeptide, sclerin, and known compound cyclosenegalin
A. Sclerin exhibited anticancer activity against human prostate carcinoma cell line
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 573
DU-145 [96]. Annona senegalensis, frequently known as wild custard apple and
wild soursop, is native to Senegal and also found in semi-arid to subhumid regions
of Africa. Most parts of the plant were used in traditional medicine. A. senegalensis
afforded annogalene, acetogenins, annosenegalin, kaurenoic acid, and (−)-roemer-
ine which indicted cytotoxic activity [97]. Further studies in organic extracts of
A. senegalensis revealed anticancer potential against HCC-1396, HEp-2, and CT-26
cancer cell lines, and highest selectivity was observed in HEP-2 cells with an IC50
value of 0.42 ± 0.09 μg/ml [98]. Methanolic extract of A. senegalensis exhibited in
vitro cytotoxic against ovarian cancer cells (A2780 with an IC50 of 28.8 μg/ml) [99].
The previous study showed methanol, butanol as well as crude extract of A. senega-
lensis showed dose - dependent anticancer activity in MDA-MB-231 cells via bind-
ing Lys −53 of the eL42 protein and preventing the crosslinking reaction with
tRNAox. Sub-fractionated extract of roots reported cytotoxicity against 293 T,
PANC-1, and HeLa with TC50 value 125.89, 211.35, and 266.07 μg/ml respec-
tively. Ent-kaurenoids reported from A. senegalensis stem bark indicated anticancer
activity against MCF-7 and PC-3 cells. Alcoholic fraction A. senegalensis was
found to possess antitumor activity against Sarcoma-180 ascites. A. senegalensis
leaf extract showed moderate cytotoxic effect against A549, HT29, MCF-7, RPMI,
and U251 cell lines. Evaluation of kaur-16-en-19-oic acid reported from the root
bark of Nigerian A. senegalensis on the pancreatic tumor (PANC-1) and Henrietta
Lacks’ cervical (HeLa) cell lines showed cytotoxic activity with an IC50 value of
0.52 and 0.74 M concentrations, respectively. Kaurenoic acid (KA) was a promising
component in the synthesis of chemotherapeutic drugs. Previous reports evidenced
apoptosis and necrosis in HL-60 cells caused by kaurene-19-oic-acid [100]. It was
also found active against human glioblastoma cell line (U87) and reported reduced
viability, and an increase in annexin V- and annexin V/PI-positive cells. Acetogenins
attributed the anticancer activity of A. senegalensis seed extract against KB and
VERO cell lines.
One of the most widely grown species Annona squamosa, also known as custard
apple, was reported to possess different pharmacological activities including antitu-
mor properties. Previous reports proved dose-dependent anticancer effect was found
in MCF-7 cell lines from silver nanoparticles made from A. squamosa leaf extract.
Silver nanoparticles made from leaf extract possessed selectivity in mammalian
cells (HEK-293) and cancerous cells (HeLa). A. squamosa leaf extract on DMBA-
induced epithelial tumor in swiss albino mice confirmed its anticancer potential.
Ethanolic extract of the A. squamosa seeds was tested for MCF-7 cells and reported
significant anticancer activity. Squamotin A-D, annosquatin IV-V, muricin O, and
squamosten B isolated from the seeds of A. squamosa were found cytotoxic in mul-
tidrug resistance cancer cells. Bistetrahydrofuran acetogenins isolated from the
ethanolic extract of A. squamosa seeds found to be showing in vitro cytotoxicity in
human cancer cell lines tested [101]. Seed extracts of A. squamosa demonstrated
DNA fragmentation and nuclear condensation and generating reactive oxygen
574 S. M. Joseph and A. R. Amala Dev
species, reducing intracellular glutathione levels, and regulating Bcl-2 and PS exter-
nalization in MCF-7 and K-562 cells. Interestingly, seed extract was found to exhibit
cytotoxicity in COLO-205 cell lines. Annotemoyin-1, annotemoyin-2, cholesteryl
glucopyranoside, and squamocin reported from seed extracts reported cytotoxic
activity. Promising anticancer constituents such assquamocin, bullatacin, uvarigran-
din A, and squamostanin A-D were reported from seed extract of A. sqamosa.
Annotemoyin-1, annotemoyin-2, squamocin, and cholesteryl glucopyranoside iso-
lated from the seeds possessed antitumor activity. Earlier investigations in A. squa-
mosa seed extract proved potent cytotoxic potential in MCF-7 and Hep G2 cells.
N-methylated lactam-containing compounds isolated from seed extracts also pos-
sessed anticancer activity. Inhibition of IL-6/Jak/Stat3 pathway was observed in a
study conducted using A. squamosa seed oil in H22 solid tumor. Fruit pericarp
extract of A. sqamosa has shown anticancer activity in Dalton’s lymphoma cells and
HeLa cells. A. squamosa bark extract possessed mono-tetrahydrofuran ring aceto-
genins found to be active against PACA-2 with a high potency of about 10–100
times than the most commonly used anticancer drug Adriamycin [102].
The following table [Table 1] comprises potent anticancer compounds reported
from different Annona species and their mode of action in various cancer cell lines.
The chemical structures of some of the isolated compounds including annonaceous
acetogenins and alkaloids are presented in the below figures [Figs. 2 & 3].
4 Conclusions
Table 1 Anticancer compounds of Annona species and their mode of action in various cancer
cell lines
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
1 A. Seeds Annomolin, Prostate tumor cell Exhibited potent
cherimola annocherimolin line (PC-3), Breast cytotoxicity
(MCF-7), and Colon
(HT-29) cancer cell
lines
Leaves Asimilobine Acute myeloid Upregulation of
leukemia cell line Bax,
downregulation of
BC-l2, and
cleavage of PARP
2 A. coriacea Roots Coriadienin, VERO and KB cell Exhibited
gigantetronenin lines anticancer activity
3 A. Seeds Sucrose octaacetate Human melanoma Exhibited potent
cornifolia (UACC-62), renel cytotoxicity
carcinoma (TK -10),
breast cancer
(MCF-7) cell line
4 A. Crude Catechin Cervical cancer cell Apoptosis via
crassiflora extract intrinsic pathway
and decreased p21
expression by
ubiquitin
proteasome
pathway
5 A. dioca Roots Squamocin, Histiocytic Exhibited
cherimolin-1, lymphoma (U-937) cytotoxicity
annonacin
Leaves Kaempferol, Ehrlich carcinoma Exhibited
3-O-[3″,6″-di-O-p- cells inhibitory effects
hydroxycinnamoyl]-
beta-galactopyranosyl-
kaempferol,
6”-O-p-
hydroxycinnamoyl-
beta-galactopyranosyl-
kaempferol
6 A. glabra Leaves Asinicin Human monocytic Mitochondria
leukemia cells mediated
(CRL-12253) anticancer and
antiproliferative
effects
Leaves Annoglacin A and B Human breast Suppressed
carcinoma (MCF-7) proliferation
and pancreatic
carcinoma
(PACA-2) cell lines
(continued)
576 S. M. Joseph and A. R. Amala Dev
Table 1 (continued)
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
Leaves Icariside D2 Human leukemia Induced apoptosis
cell line (HL-60) and decreased
phosphorylation of
AKT in cells
Fruits Annoglabasin H Lung Exhibited
adenocarcinoma cell significant
line (LU-1), human cytotoxic activity
breast carcinoma
(MCF-7), human
melanoma
(SK-Mel2), KB
Seeds Bullatacin CEM/VLB cell line Induced apoptosis,
necrosis and
upregulated the
expression of
cyclin kinase
inhibitor (WAF1/
p21). Induced cell
cycle arrest at the
G0/G1 phase
Fruits Annoglabayin Human liver cancer Apoptosis via
cell line (Hep G2) mitochondrial
pathway
Fruits Cunabic acid and Liver cancer (HLC) Apoptosis via
ent-kauran-19-al-17-oic cell line downregulation of
acid SMMC-7721 BCL-2 gene and
upregulation of bax
gene
7 A. Stem Actinodaphnine, Breast and colon Inhibited tumor
hypoglauca anonaine, isoboldine, cancer cells. formation
nornuciferine
8 A. jahnii Twigs Annodienin, Human solid Cytotoxicity
jahnonacin, annojahnin cancerous cell lines
9 A. montana Leaves Montanacin F Lewis lung Exhibited cytotoxic
carcinoma (LLC) activity
tumor cell lines
Seeds Montacin and Human ovarian Exhibited cytotoxic
cis-Montacin cancer cell line activity
(1A9)
Seeds Montalicins G and H, Human hepatoma Exhibited cytotoxic
monlicins A and B, cells (Hep G2) activity
(+)-monhexocin and
(−)-monhexocin
murisolin
4-deoxyannomo-ntacin,
muricatacin
(continued)
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 577
Table 1 (continued)
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
10 A. muricata Leaves Muricatocins A and B Lung tumor cell lineEnhanced cytotoxic
(A-549) activity
Leaves Muricoreacin, Colon cancer cell Upregulation of
murihexocin (HT-29, HCT-116) Bax,
downregulation of
Bcl-2 proteins and
activated initiator
and executioner
Caspases
Leaves Annomutacin A-549 cells of lungs Apoptosis by
promoting
mitochondria
mediated signalling
pathway through
inhibiting NF-kB
factor
Leaves (E)-Caryophyllene, Breast cancer cell Exhibited cytotoxic
eugenol (MCF-7) effect
Leaves Annomuricine Rat breast cancer Reduced
cell proliferative
indexes
Leaves Annopentocin BPH-1 cells Exerted
antiproliferative
effect via
upregulation of bax
gene and
downregulation of
Bcl-2
Leaves Annomuricine, Pancreatic Exhibited
muricapentocin carcinoma repressive effect
(PACA-2) and colon
adenocarcinoma
(HT-29) cell
Fruits Muricin M and muricin Prostate cancer Exhibited
N (PC-3) cells cytotoxicity
11 A. purpurea Roots Annopurpurici-ns A–D HeLa and HepG2 Mitochondrial
cells membrane
depolarization and
apoptosis
12 A. reticulata Leaves Annomonicin Colon cancer Exhibited cytotoxic
(HCT15), Human effect
lung cancer
(Hop65), and
Human hepatoma
(Hep G2) cell lines
(continued)
578 S. M. Joseph and A. R. Amala Dev
Table 1 (continued)
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
Leaves Rolliniastatin Breast cancer cell Caspases
(T-47D) dependent
apoptosis.
Downregulated
Bcl-2 and
upregulated Bax,
Bak
Seeds Annonacin T-24 Bladder cancer Bax expression was
cells induced, caspase-3
activity enhanced
and caused
apoptosis
Seeds Bullatacin Leukemia cell line Cell death via
(K-562) and breast apoptosis
cancer cell line
(MCF-7)
Fruits Catechin Breast cancer cell Inhibition via
line (MCF-7) apoptosis
13 A. Seeds Sclerin, cyclosenegalin Human prostate Exhibited
scleroderma A carcinoma cell line anticancer activity
DU-145
14 A. Leaves (-)-Roemerine Breast cancer Exhibited
senegalensis MDA-MB-231 cells dose-dependent
cytotoxicity via
targeting the
ribosomal protein
eL42 and arresting
the crosslinking
reaction with
tRNAox
Stem Ent-kaurenoids Breast cancer Exhibited
bark (MCF-7) cells, cytotoxicity
prostate cancer
(PC-3) cells
Bark Kaurenoic acid Human embryonic Exhibited
kidney cells cytotoxicity
expressing SV40
Large T-antigen
(293 T), pancreatic
tumor (PANC-1)
cell lines and
henrietta lacks’
cervical cancer cell
line (HeLa)
15 A. Leaves Annoreticuin Breast cancer cell Induced Apoptosis
squamosa (MCF-7)
(continued)
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 579
Table 1 (continued)
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
Seeds Annosquatin A, B Human breast Induction of
cancer cell line reactive oxygen
(MCF-7), human species generation,
leukemia cell line reduced
(K-562), human intracellular
colon carcinoma glutathione levels
(COLO-205) and regulation of
Bcl-2 and PS
externalization
Seeds (-)-Anonaine H-22 solid tumor Inhibition of IL-6/
cell Jak/Stat3 pathway
Fruit (-)-Ent-kaur-16-en-19- Dalton’s lymphoma Exhibited cytotoxic
oic acid, 16α,17- cells, HeLa cells activity
dihydroxy-ent-kauran-
19-oic acid
Seeds Asimilobine Human colon cancer Increased
cell (WiDr) expression of
caspase-3, induced
G2/M phase and
apoptosis
Seeds Squamoxinone-D Nasopharyngeal Inhibited
cancer (KB) cells, proliferation
breast cancer
(MCF- 7) cells, lung
cancer (A-549)
cells, leukemic
(K-562) cells
Seeds Dieporeticenin B, SMMC 7721/T, Exerted inhibitory
squamocin, annosquatin MCF-7/ADR activity
III
Seeds Squadiolins A, B Hep G2 hepatoma Exhibited
cells, significant
MDA-MB-231 cytotoxic activity
breast cancer cells
Seeds Squafosacin B Human Hep G2, 3B Exhibited
Hepatoma and significant
Breast cancer cells cytotoxic activity
(MCF-7)
Seeds Annosquatin A, MCF-7, A-549 cell Exhibited high
annosquatin B lines cytotoxic
selectivity
(continued)
580 S. M. Joseph and A. R. Amala Dev
Table 1 (continued)
Sl. Annona Plant Animal model / Cell
No. Species part Components identified line Mode of action
Seeds Squamocin - I, II, III, Hep G2, SMMC Exhibited cytotoxic
squamoxinone-D 7721, human activity
hepatocellular
carcinoma cell line
(BEL 7402), gastric
cancer cells (BGC
803), lung cancer
cells (H460)
Bark Coclaurine DMBA painted Enhanced lipid
hamsters peroxidation
References
1. Alali FQ, Liu XX, McLaughlin JL (1999) Annonaceous acetogenins: recent progress. J Nat
Prod 62:504–540. https://doi.org/10.1021/np980406d
2. Oberlies NH, Jones JL, Corbett TH, Fotopoulos SS, McLaughlin JL (1995) Tumor cell
growth inhibition by several annonaceous acetogenins in an in vitro disk diffusion assay.
Cancer Lett 96:55–62. https://doi.org/10.1016/0304-3835(95)92759-7
3. Quílez AM, Fernandez Arche MA, García Giménez MD, De La Puert R (2018) Potential
therapeutic applications of the genus Annona: local and traditional uses and pharmacology. J
Ethnopharmacol 225:244–270. https://doi.org/10.1016/j.jep.2018.06.014
4. Jacobo Herrera N, Pérez Plasencia C, Castro-Torres VA, Martínez Vázquez M, González
Esquinca AR, Zentella DA (2019) Selective acetogenins and their potential as anticancer
agents. Front Pharmacol 10:783. https://doi.org/10.3389/fphar.2019.00783
5. Zafra Polo MC, Gonzalez MC, Estornell E, Sahpaz S, Cortes D (1996) Acetogenins from
Annonaceae, inhibitors of mitochondrial complex I. Phytochemistry 42:253–271. https://doi.
org/10.1016/0031-9422(95)00836-5
6. Elhawary SS, Tantawy MEE, Rabeh MA, Fawaz NE (2013) DNA fingerprinting, chemical
composition, antitumor and antimicrobial activities of the essential oils and extractives of
four Annona species from Egypt. J Nat Sci Res 3:59–68
7. Chen CY, Chang FR, Chiu HF, Wu MJ, Wu YC (1999) Aromin-A, an annonaceous ace-
togenin from Annona cherimola. Phytochemistry 51:429–433. https://doi.org/10.1016/
s0031-9422(99)00002-3
8. Haykal T, Nasr P, Hodroj MH, Taleb RI, Sarkis R, Moujabber MNE, Rizk S (2019) Annona
cherimola seed extract activates extrinsic and intrinsic apoptotic pathways in leukemic cells.
Toxins 11:506. https://doi.org/10.3390/toxins11090506
9. Jyothi BA, Venkatesh KP, Chakrapani RAR (2011) Phytochemical and pharmacological
potential of Annona cherimola - A review. Int J Phytomedicine 3:439–447
10. Kim DH, Ma ES, Suk KD, Son JK, Lee JS, Woo MH (2001) Annomolin and annocheri-
molin, new cytotoxic annonaceous acetogenins from Annona cherimola seeds. J Nat Prod
64:502–506. https://doi.org/10.1021/np000335u
11. Ammoury C, Younes M, Khoury ME, Haykal T, Nasr P, Sily M, Taleb RI, Sarkis R, Khalife
R, Rizk S (2009) The pro-apoptotic effect of a terpene-rich Annona cherimola leaf extract
on leukemic cell lines BMC. Complement Altern Med 19:365. https://doi.org/10.1186/
s12906-019-2768-1
12. Macuer-Guzmán J, Bernal G, Jamett-Díaz F, Ramírez-Rivera S, Ibanez C (2019) Selective
and apoptotic action of ethanol extract of Annona cherimola seeds against human stom-
ach gastric adenocarcinoma cell line AGS. Plant Foods Hum Nutr 74:322–327. https://doi.
org/10.1007/s11130-019-00742-w
13. Cortes D, Myint SH, Hocquemiller R (1991) Molvizarin and motrilin two novel cytotoxic
bistetrahydrofuranic gamma lactone acetogenins from Annona cherimola. Tetrahedron
47:8195–8202. https://doi.org/10.1016/S0040-4020(01)91014-2
14. Woo MH, Chung SO, Kim DH (1999) Cis-Annonacin and (2,4)-cis-and trans-Isoannonacins:
cytotoxic monotetrahydrofuran annonaceous acetogenins from the seeds of Annona cheri-
mola. Arch Pharm Res 22:524–528. https://doi.org/10.1007/bf02979164
15. Cortes D, Myint SH, Dupont B, Davoust D (1993) Bioactive acetogenins from seeds of Annona
cherimola. Phytochemistry 32:1475–1482. https://doi.org/10.1016/0031-9422(93)85162-k
16. Gomes INF, Silva-Oliveira RJ, Oliveira Silva VA, Rosa MN, Vital PS, Barbosa MCS, dos
Santos FV, Junqueira JGM, Severino VGP, B. G. Oliveira, W. Romao, R.M. Reis, de de
Azambuja Ribeiro RIM. Annona coriacea Mart. fractions promote cell cycle arrest and
inhibit autophagic flux in human cervical cancer cell lines. Molecules. 2019;24:3963
17. Roblot JF, Mahuteau A, Cavé A (1996) Coriadienin. The first annonaceous acetogenin with
two double bonds isolated from Annona coriaceae. J Nat Prod 59:528–530
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 583
18. Lima LARS, Alves TMA, Zani CL, Santos LP, Boaventura MAD (2013) Cytotoxic potential
of sucrose octaacetate from the ethanol extract of the seeds of Annona cornifolia A. St.-Hil.
(Annonaceae). Sucrose: Properties, Biosynthesis and Health Implications:143–154
19. Lima LARS, Alves TMA, Zani CL, Santos LP, Pimenta BMAD (2012) Antioxidant and cyto-
toxic potential of fatty acid methyl esters from the seeds of Annona cornifolia A. St.-Hil.
(Annonaceae). Food Res Int 48:873–875
20. Arruda HS, Pastore GM (2019) Araticum (Annona crassiflora Mart.) as a source of nutrients
and bioactive compounds for food and non-food purposes: A comprehensive review. Food
Res Int 123:450–480. https://doi.org/10.1016/j.foodres.2019.05.011
21. Rocha RS, Kassuya CAL, Formagio ASN, Mariana de Oliveira Mauro M, Andrade-Silva
MACD, Cunha-Laura AL, Vieira MC, Oliveira RJ (2016) Analysis of the anti-inflammatory
and chemopreventive potential and description of the antimutagenic mode of action of
the Annona crassiflora methanolic extract. Pharm Biol 54:35–47. https://doi.org/10.310
9/13880209.2015.1014567
22. Silva VAO, Alves ALV, Rosa MN, Silva LRV, Melendez FP, Cury FP, Gomes INF, Tansini
A, Longato GB, Martinho O, Oliveira BG, Pinto FE, Romao W, Ribeiro RIMA, Reis RM
(2018) Hexane partition from Annona crassiflora Mart. Promotes cytotoxity and apoptosis on
human cervical cancer cell lines. Investig New Drugs 37:602–615. https://doi.org/10.1007/
s10637-018-0657-y
23. Pimenta LPS, Mendonça DD, Pretti DL, Cruz BLD, Leite EA, DeOliveira MC (2011)
Evaluation of in vivo antitumor activity of Annona crassiflora wood extract. Int J Pharm Sci
Drug Res 3:270–273. https://doi.org/10.1080/14786419.2014.900496
24. Prado LG, Arruda HS, Araujo NMP, Braga LEO, Banzato TP, Pereira GA, Figueiredo MC,
GoisRuiz ALT, Eberlinf MN, Carvalho JE, Vendramini-Costa DB, MariaPastore GM (2020)
Antioxidant, antiproliferative and healing properties of araticum (Annona crassiflora Mart.)
peel and seed. Food Res Int 133:109168. https://doi.org/10.1016/j.foodres.2020.109168
25. Kanashiro MM, Paes MM, Vega MRG, Cortes D, Cabedo N (2015) Cytotoxic activities of
acetogenins from Annona dioica roots. Int J Mol Sci 4:19–29
26. Vega MRG, Esteves-Souza A, Vieira IJC, Mathias L, Braz-Filho R, Echevarria A (2007)
Flavonoids from Annona dioica leaves and their effects in Ehrlich carcinoma cells,
DNA-topoisomerase I and II. J Am Chem Soc 18:1554–1559. https://doi.org/10.1590/
S0103-50532007000800016
27. Formagio AS, Kassuya CA, Neto FF, Volobuff CR, Iriguchi EK, Vieira Maria do C, Foglio
MA (2013) The flavonoid content and antiproliferative, hypoglycaemic, anti-inflammatory
and free radical scavenging activities of Annona dioica St. Hill. BMC Complement Altern
Med 13:14. https://doi.org/10.1186/1472-6882-13-14
28. Liu XX, Alali FQ, Pilarinou E, McLaughlin JL (1999) Two bioactive mono-tetrahydrofuran
acetogenins, annoglacins A and B, from Annona glabra. Phytochemistry 50:815–821. https://
doi.org/10.1016/s0031-9422(98)00466-x
29. Hien NTT, Nhiem NX, Yen DTH, Hang DT, Tai BH, Quang TH, Anh HLT, Kiem PV, Minh
CV, Kim EJ, Kim SH, Kang HK, Kim YH (2015) Chemical constituents of the Annona gla-
bra fruit and their cytotoxic activity. Pharm Biol 53:1602. https://doi.org/10.3109/1388020
9.2014.993042
30. Abdel-lateff A, El-Menshawi BS, Hagag MY, Nawwar MA (2009) Cytotoxic acetogenins
from Annona glabra cultivated in Egypt. Pharm Res 1:130–135
31. Anh Hle T, Hien NT, Hang DT, Ha TM, Nhiem NX, Hien TT, Thu VK, Thao do T, Van Minh
C, Kiem PV (2014) Ent-kaurane diterpenes from Annona glabra and their cytotoxic activi-
ties. Nat Prod Commun 9:1681–1682
32. Cochrane CB, Nair PK, Melnick SJ, Resek AP, Ramachandran C (2008) Anticancer effects
of Annona glabra plant extracts in human leukemia cell lines. Anticancer Res 28:965–971
33. Padmaja V, Thankamany V, Hara N, Fujimoto YY, Hisham A (1995) Biological activities of
Annona glabra. J Ethnopharmacol 48:21–24. https://doi.org/10.1016/0378-8741(95)01277-k
584 S. M. Joseph and A. R. Amala Dev
34. Chen CH, Hsieh TJ, Liu TZ, Chern CL, Hsieh PY, Chen CY (2004) Annoglabayin, a novel
dimeric kaurane diterpenoid and apoptosis in Hep G2 cells of annomontacin from the fruits
of Annona glabra. J Nat Prod 67:1942–1946. https://doi.org/10.1021/np040078j
35. Zhang YH, Peng HY, Xia GH, Wang MY (2004) Anticancer effect of two diterpenoid com-
pounds isolated from Annona glabra Linn. Acta Pharmacol Sin 25:937–942
36. Rinaldi MVN, Díaz IEC, Suffredini IB, Moreno PRM (2017) Alkaloids and biological activ-
ity of beribá (Annona hypoglauca). Rev Bras 27:77
37. dos Santos RC, Chagas EA, de Melo Filho AA, Takahashi JA, Montero IF, Chagas PC,
Ribeiro PRE, de Melo ACGR (2018) Bioactive extracts from Annona hypoglauca. Chem
Eng Trans 64
38. Colman-saizarbitoria T, Liu X, Craig Hopp D, Johnson HA, Alali FQ, Rogers LL, McLaughlin
JL (1999) Annodienin and Jahnonacin: new bioactive nontetrahydrofuran annonaceous ace-
togenins from twigs of Annona jahnii. J Nat Prod 14:65
39. Colman-Saizarbitoria T, Johnson HA, Alali FQ, Hopp DC, Rogers LL, McLaughlin JL
(1998) Annojahnin from Annona Jahnii: A possible precursor of mono-tetrahydrofuran ace-
togenins. Phytochemistry 49:1609–1616
40. Wang LQ, Min BS, Li Y, Nakamura N, Qin GW, Li CJ, Hattori M (2002) Annonaceous
acetogenins from the leaves of Annona montana. Bioorg Med Chem 10:561–565. https://doi.
org/10.1016/s0968-0896(01)00303-0
41. Wang LQ, Li Y, Min BS, Nakamura N, Qin GW, Li CJ, Hattori M (2001) Cytotoxic mono-
tetrahydrofuran ring acetogenins from leaves of Annona montana. Planta Med 67:847–852
42. Bailon-Moscoso N, Benavides JCR, Orellana MIR, Ojeda K, Granda G, Ratoviski EA (2016)
Ostrosky- Wegman P. cytotoxic and genotoxic effects of extracts from Annona montana fruit.
Food Agric Immunol 27:559–569. https://doi.org/10.1080/09540105.2016.1148121
43. Liaw CC, Chang FR, Wu YC, Wang HK, Nakanishi Y, Bastow KF, Lee KH (2004) Montacin
and cis-montacin, two new cytotoxic mono tetrahydrofuran annonaceous acetogenins from
Annona montana. J Nat Prod 67:1804–1808. https://doi.org/10.1021/np030216p
44. Liaw CC, Chang FR, Chen SL, Wu CC, Lee KH, Wu YC (2005) Novel cytotoxic mono-
tetrahydrofuranic annonaceous acetogenins from Annona montana. Bioorg Med Chem
13:4767–4776
45. Wu TS, Jong TT, Tien HJ, Kuoh CS, Furukawa H, Lee KH (1987) Annoquinone-A, an anti-
microbial and cytotoxic principle from Annona montana. Phytochemistry 26:1623–1625.
https://doi.org/10.1016/S0031-9422(00)82257-8
46. Moghadamtousi SZ, Fadaeinasab M, Nikzad S, Mohan G, Ali HM, Kadir HA (2015) Annona
muricata (Annonaceae): A review of its traditional uses, isolated acetogenins and biological
activities. Int J Mol Sci 16:15625–15658. https://doi.org/10.3390/ijms160715625
47. Prasad Shashanka K, Varsha V, Devegowda D (2019) Anti-cancer properties of Annona
muricata (L.): A review, Medicinal Plants. Int J Phytomed Relat Ind 11:123. https://doi.
org/10.5958/0975-6892.2019.00016.9
48. Dilipkumar JP, Agliandeshwari D (2017) Preparation & evaluation of Annona muricata
extract against cancer cells with modified release. Pharma Tutor 5:63–106
49. Abdul Wahab SM, Jantan I, Haque MA, Arshad L (2018) Exploring the leaves of Annona
muricata as a source of potential anti-inflammatory and anticancer agents. Front Pharmacol
9. https://doi.org/10.3389/fphar.2018.00661
50. Moghadamtousi KHA, Paydar M, Rouhollahi E, Karimian H (2014) Annona muri-
cata leaves induced apoptosis in A549 cells through mitochondrial-mediated pathway
and involvement of NF-κB. BMC Complement Altern Med 14:299–302. https://doi.
org/10.1186/1472-6882-14-299
51. Patrikios I, Stephanou A, Andreas Y (2015) Graviola: A systematic review on its anticancer
properties. Asian Pac J Cancer Prev 3:128–131. https://doi.org/10.11648/j.ijpc.20170306.14
52. Qazi AK, Siddiqui JA, Jahan R, Chaudhary S, Walker LA, Sayed Z, Jones DT, Batra SK,
Macha MA (2018) Emerging therapeutic potential of graviola and its constituents in cancers.
Carcinogenesis 39:522–533. https://doi.org/10.1093/2Fcarcin/2Fbgy024
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 585
53. Wu FE, Zeng L, Gu ZM, Zhao GX, Zhang Y, Schwedler JT, McLaughlin JL, Sastrodihardjo
S (1995) Muricatocins A and B, two new bioactive monotetrahydrofuran annonaceous aceto-
genins from the leaves of Annona muricata. J Nat Prod 58:902–908. https://doi.org/10.1021/
np50120a013
54. Daud NM, Yaakob H, Rosdi M (2016) Acetogenins of Annona muricata leaves: charac-
terization and potential anticancer study. Integr Cancer Sci Ther 3:543–551. https://doi.
org/10.15761/icst.1000202
55. Syed Najmuddin SUF, Romli MF, Hamid M, Alitheen NB, Afizan NM, Rahman NA (2016)
Anti-cancer effect of Annona muricata Linn leaves crude extract (AMCE) on breast cancer
cell line. BMC Complement Altern Med 16:311. https://doi.org/10.1186/s12906-016-1290-y
56. Growther L (2018) Anticancer activity of Annona muricata leaf extracts and screening of
bioactive phytochemicals. Int J Pharm Biol Sci 201(8):475–481
57. Moghadamtousi MZ, Karimian H, Rouhollahi E, Paydar M, Fadaeinasab M, Kadir HA (2014)
Annona muricata leaves induce G1 cell cycle arrest and apoptosis through mitochondria-
mediated pathway in human HCT-116 and HT-29 colon cancer cells. J Ethnopharmacol
156:277–289. https://doi.org/10.1016/j.jep.2014.08.011
58. Moghadamtousi SZ, Kadir HA, Paydar M, Rouhollahi E, Karimian H (2014) Annona
muricata leaves induced apoptosis in A549 cells through mitochondrial-mediated path-
way and involvement of NF-Κb. BMC Complement Altern Med 14:299. https://doi.
org/10.1186/1472-6882-14-299
59. Torres MP, Rachagani S, Purohit V, Pandey P, Joshi S, Moore ED, Johansson SL, Singh PK,
Ganti AK, Batra SK (2012) Graviola: A novel promising natural-derived drug that inhibits
tumorigenicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering
cell metabolism. Cancer Lett 323:29–40. https://doi.org/10.1016/j.canlet.2012.03.031
60. Owolabi MS, Ogundajo AL, Dosoky NS, Setzer WN (2013) The cytotoxic activity of Annona
muricata leaf oil from Badagary. Nigeria Am J Essent Oil 1:1–3
61. Wu FE, Gu ZM, Zeng L, Zhao GX, Zhang Y, McLaughlin JL, Sastrodihardjo S (1995) Two
new cytotoxic monotetrahydrofuran annonaceous acetogenins, annomuricins A and B from
the leaves of Annona muricata. J Nat Prod 58:830–836. https://doi.org/10.1021/np50120a002
62. Hamizah S, Roslida AH, Fezah O, Tan KL, Tor YS, Tan CI (2012) Chemo preventive poten-
tial of Annona muricata L leaves on chemically-induced skin papillomagenesis in mice.
Asian Pac J Cancer Prev 13:2533–2539. https://doi.org/10.7314/apjcp.2012.13.6.2533
63. Sulistyoningrum E, Rachmani EPN, Baroroh HN, Rujito L (2017) Annona muricata leaves
extract reduce proliferative indexes and improve histological changes in rat's breast cancer. J
App Pharm Sci 7:149–155. https://doi.org/10.7324/japs.2017.70120
64. Gavamukulya Y, Abou-Elella F, Wamunyokoli F, AEl-Shemy H. (2014) Phytochemical
screening, anti-oxidant activity and in vitro anticancer potential of ethanolic and water
leaves extracts of Annona muricata (Graviola). Asian Pac J Trop Med 7:355–363. https://doi.
org/10.1016/S1995-7645(14)60258-3
65. Astirin O, Artanti A, Fitria M, Perwitasari E, Prayitno A (2013) Annona muricata Linn leaf
induces apoptosis in cancer cause virus. J Cancer Ther 4:1244–1250. https://doi.org/10.4236/
jct.2013.47146
66. Yang H, Liu N, Lee SC (2016) Ethanol extract of Annona muricata. L induces liver cancer cell
apoptosis through ROS pathway. Biomed Pharmacol J 9:919–925. https://doi.org/10.13005/
bpj/1030
67. Rahman HA, Wan-Ibrahim WS, Ismail N, Ismail TN, Mohd-Salleh SF, Pak-Kai Wong M,
Samad MA, Hashim MM (2018) Phytocompounds of Annona muricata leaves extract and
cytotoxic effects on breast cancer cells. Asian Pac J Trop Med 11:659–665. https://doi.
org/10.4103/1995-7645.248337
68. Muhartono A, Sutyarso S, Kanedi M (2016) Anti-proliferative and apoptotic effects of
mucoxin (acetogenin) in T47D breast cancer cells. Biomed Pharmacol J. 9:491–498. https://
doi.org/10.13005/bpj/963
586 S. M. Joseph and A. R. Amala Dev
69. Rosdi M, Daud NM, Zulkifli RM, Yaakob H (2015) Cytotoxic effect of Annona muricata
Linn leaves extract on capan-1 cells. J Appl Pharm Sci 5:45–48. https://doi.org/10.7324/
japs.2015.50508
70. Meenakshisundaram S, Krishnamoorthy V, Jagadeesan Y, Vilwanathan R, Balaiah A (2020)
Annona muricata assisted biogenic synthesis of silver nanoparticles regulates cell cycle arrest
in NSCLC cell lines. Bioorg Chem 95:103451. https://doi.org/10.1016/j.bioorg.2019.103451
71. Asare GA, Afriyie D, Ngala RA, Abutiate H, Doku D, Mahmood SA, H. Rahman
H. (2015) Antiproliferative activity of aqueous leaf extract of Annona muricata L. on the
prostate, BPH-1 cells, and some target genes. Integr Cancer Ther 14:65–74. https://doi.
org/10.1177/1534735414550198
72. Kim GS, Zeng L, Alali F, Rogers LL, Wu FE, McLaughlin JL, Sastrodihardjo S (1998) Two
new mono-tetrahydrofuran ring acetogenins, annomuricin E and muricapentocin from the
leaves of Annona muricata. J Nat Prod 61:432–436. https://doi.org/10.1021/np970534m
73. Sun S, Liu J, Zhou N, Zhu W, Dou QP, Zhou K (2016) Isolation of three new annonaceous
acetogenins from graviola fruit (Annona muricata) and their anti-proliferation on human
prostate cancer cell PC-3. Bioorg Med Chem Lett 26:4382–4385. https://doi.org/10.1016/j.
bmcl.2015.06.038
74. Rieser MJ, Gu ZM, Fang XP, Zeng L, Wood KV, McLaughlin JL (1996) Five novel mono-
tetrahydrofuran ring acetogenins from the seeds of Annona muricata. J Nat Prod 59:100–108.
https://doi.org/10.1021/np960037q
75. Li DY, Yu JG, Zhu JX, Yu DL, Luo XZ, Sun L, Yang SL (2001) Annonaceous aceto-
genins of the seeds from Annona muricata. J Asian Nat Prod Res 3:267–276. https://doi.
org/10.1080/10286020108040366
76. Sonnet PE, Jacobson M (1971) Tumor inhibitors II: cytotoxic alkaloids from Annona pur-
purea. J Pharm Sci 60:1254–1125. https://doi.org/10.1002/jps.2600600835
77. Hernandez-Fuentes GA, García-Argaez AN, Peraza Campos AL, Delgado-Enciso I, Muniz-
Valencia R, Martínez-Martínez FJ, Toninello A, Sandoval ZG, Mojica-Sánchez JP, Via LD,
Parra-Delgado H (2019) Cytotoxic acetogenins from the roots of Annona purpurea. Int J Mol
Sci 20:1870. https://doi.org/10.3390/ijms20081870
78. Chávez D, Mata R (1999) Purpuracenin: A new cytotoxic adjacent bis-tetrahydrofuran
annonaceous acetogenin from the seeds of Annona purpurea. Phytochemistry 50:823–828.
https://doi.org/10.1016/s0031-9422(98)00553-6
79. Chávez D (1998) R. Mata R. Purpurediolin and purpurenin, two new cytotoxic adjacent bis-
tetrahydrofuran annonaceous acetogenins from the seeds of Annona purpurea. J Nat Prod
61:580–584. https://doi.org/10.1021/np970410
80. Hernandez-Fuentes GA, Peraza Campos AL, Silvia G (2020) Ceballos-Magana, Roberto
Muniz-Valencia, Hortensia Parra-Delgado, HPLC-DAD method for the detection of five
annopurpuricins in root samples of Annona purpurea. Phytochem Anal 31:472–479. https://
doi.org/10.1002/pca.2910
81. Pathak K, Zaman MK (2014) An overview on medicinally important plant - Annona reticu-
lata Linn. Int J Pharmacogn Phytochem Res 5:299–301
82. Shivanna LM, Urooj AM (2019) Apoptotic effects of Annona reticulata leaves extract in
HT-29 cell lines. Asian J Biol Sci 12:820–831. https://doi.org/10.3923/ajbs.2019.820.831
83. Rajarajan AP, Senthil KR (2016) Assessment of antineoplastic potential of Annona reticulata
Linn. On human cancer cells. Asian J Pharm Clin Res 9:408–411
84. Chang FR, Wu YC, Duh CY, Wang SK (1993) Studies on the acetogenins of formosan annona-
ceous plants. II. Cytotoxic acetogenins from Annona reticulata. J Nat Prod 56:1688–1694.
https://doi.org/10.1021/np50100a005
85. Jamkhande PG, A.S. Wattamwar AS. (2015) Annona reticulata Linn. (bullock's heart):
plant profile, phytochemistry and pharmacological properties. J Tradit Complement Med
5:144–152. https://doi.org/10.1016/j.jtcme.2015.04.001
86. Gingine AP, Mandge SV, Jamkhande PG (2016) In vitro evaluation of anticancer activity of
methnaolic extract of Annona reticulata Linn. (Ramphal) leaves on different human cancer
cell lines. J Anal Pharm Res 2016 3. https://doi.org/10.15406/japlr.2016.03.00087
Annonaceae: Tropical Medicinal Plants with Potential Anticancer Acetogenins… 587
87. Mondal SK, Mondal NB, Mazumder UK (2007) In vitro cytotoxic and human recombi-
nant caspase inhibitory effect of Annona reticulata leaves. Indian J Pharmacol 39:253–254.
https://doi.org/10.4103/0253-7613.37279
88. Roham PH, Kharat KR, Mungde P, Jadhav MA, Surinder JM (2016) Induction of mito-
chondria mediated apoptosis in human breast cancer cells (T-47D) by Annona reticulata
L. leaves methanolic extracts. Nutr Cancer 68:305–311. https://doi.org/10.1080/0163558
1.2016.1142583
89. Yuan SS, Chang HL, Chen HW, Yeh YT, Kao YH, Lin KH, Wu YC, Su JH (2003) Annonacin,
a mono-tetrahydrofuran acetogenin, arrests cancer cells at the G1 phase and causes
cytotoxicity in a bax- and caspase-3-related pathway. Life Science 72:2853–2861. https://
doi.org/10.1016/s0024-3205(03)00190-5
90. Yogananth Y (2018) Evaluation of antitumor properties of Annona reticulata (L) seeds on
cancer cell lines. J Pharma Res 7:132–135. https://doi.org/10.5281/zenodo.1308605
91. Suresh HM, Shivakumar B, Hemalatha K, Heroor SS, Hugar DS, KRS SR (2011) In vitro
antiproliferative activity of Annona reticulata roots on human cancer cell lines. Pharm Res
3:9–12. https://doi.org/10.4103/0974-8490.79109
92. Suresh HM, Shivakumar B, Shivakumar SI (2011) Inhibitory potential of the ethanol extract
of Annona reticulata Linn. against melanoma tumor. J Nat Prod 2:168–172. https://doi.
org/10.4103/2229-5119.92846
93. Rayar A, Manivannan R (2016) Antioxidant and anticancer activities of (+)-catechin isolated
from Annona reticulata Linn. Int J Recent Sci Res 7:9451–9456
94. Queiroz EF, Roblot F, Cavé A, Hocquemiller R, Serani L, Laprévote O, de Paulo MA
(1999) New bistetrahydrofuran acetogenin from the roots of Annona salzmanii. J Nat Prod
62:710–713
95. Costa EV, Dutra LM, Salvador MJ, Ribeiro LHG, Gadelha FR (2013) Ernesto de Carvalho
J. chemical composition of the essential oils of Annona pickelii and Annona salzmannii
(Annonaceae) and their antitumor and trypanocidal activities. Nat Prod Res 27:997–1001
96. Cen-Pacheco F, Valerio-Alfaro G, Santos-Luna D, Fernández J (2019) Sclerin, a new cyto-
toxic cyclononapeptide from Annona scleroderma. Molecules 24:554
97. Okhale SE, Akpan E, Fatokun OT, Esievo KB, Kunle OF (2016) Annona senegalensis
Persoon (Annonaceae): A review of its ethnomedicinal uses, biological activities and phyto-
compounds. J Pharmacogn Phytochem 5:211–219
98. Biseko EZ, Swai HS, Mbugua RW, Ndungu JW, Chepng’etich JC, Gathirwa JW (2019) In
vitro antiproliferative potential of Annona senegalensis Pers. and Allophylus africanus P
Beauv plant extracts against selected cancer cell lines. J Med Plants Res 13:304–311. https://
doi.org/10.5897/jmpr2019.6785
99. Ajaiyeoba EO, Falade MO, Ogbole O, Okpako LC (2006) In vivo antimalarial and cytotoxic
properties of Annona senegalensis extract. Afr J Tradit Complement Altern Med 3:137–141
100. Cavalcanti BC, Bezerra DP, Magalhães HIF, Moraes MO, Lima MAS, Silveira ER, Câmara
CAG, Rao VS, Pessoa C, Costa-Lotufo LV (2009) Kauren-19-oic acid induces DNA dam-
age followed by apoptosis in human leukemia cells. J Appl Toxicol 29:560–568. https://doi.
org/10.1002/jat.1439
101. Chen Y, Chen JW, Li X (2011) Cytotoxic bistetrahydrofuran annonaceous acetogenins from
the seeds of Annona squamosa. J Nat Prod 74:2477–2481. https://doi.org/10.1021/np200708q
102. Hopp DC, Zeng L, Gu ZM, Kozlowski JK, McLaughlin JL (1997) Novel mono-
tetrahydrofuran ring acetogenins, from the bark of Annona squamosa, showing cytotoxic
selectivities for the human pancreatic carcinoma cell line, PACA-2. J Nat Prod 60:581–586.
https://doi.org/10.1021/np9701283
Biocolorant from Anisochilus carnosus:
A Natural Food Preservative
1 Introduction
Color plays a significant role in the consumer’s acceptance of food products. Since
color is associated with flavor, it affects the food quality perception and stimulates
appetite. The variety of naturally derived colors are often used to enhance the
appearance and acceptability of foods such as annatto, paprika, indigo, cochineal,
and turmeric [1]. Generally, food colorants have been added to many foods to make
them more appealing and acceptable. Synthetic food colors come in a variety of
color shades, but many studies have carried out on the toxicity of synthetic food
colors. Toxic and carcinogenic properties of approved synthetic colors have been
reported. The use of large number of synthetic colors pollutes the environment,
disrupts the ecological balance, and rises many health risks [2].
Natural colors also act as a potent antioxidant when added to foods. Plant anti-
oxidants can be classified into three categories: phenolic compounds, carotenoids,
and vitamins. In addition to being the most abundant plant compounds with antioxi-
dant activity, certain phenolic compounds often have antimicrobial and antifungal
properties, as well as significant effects on flavors and textures of foods. Some natu-
ral pigments like β-carotene, lycopene, quercetin, and rutin have antioxidant activ-
ity which are also used as food colorants [3]. Many compounds can inhibit oxidation,
but due to safety concerns, a few are suitable for human consumption and generally
Suman Thamburaj, Chayanika Sarma, Anju Mariam Johnson and Akhila Etikala contributed
equally with all other contributors.
recognized as safe food grade antioxidants approved by regulatory bodies that are
applied to foods.
Synthetic phenolic antioxidants with antimicrobial action include butylated
hydroxyanisole (BHA), butylated hydroxytoluene (BHT), and monotertiary butyl-
hydroquinone (TBHQ). The presence of a hydroxyl group on the molecule, the
compound’s lipid solubility, and the degree of steric hindrance appear to influence
the antibacterial action of phenolic antioxidants. The antibacterial action of pheno-
lic antioxidants in foods has been studied against bacterial growth and by-products
(gram positive and negative, spore and nonspore formers, spoilage and pathogenic),
moulds, and yeasts. The concentration of phenolic antioxidants with antibacterial
action in food products ranged from 30 to 10,000 ppm. The mechanism of inhibition
by phenolic antioxidants has been discovered to alter the function and composition
of the cellular membrane, DNA, RNA, protein, and lipid production, and mitochon-
drion function.
Phenolic antioxidants are insoluble in water, but soluble in organic solvents such
as ethanol and propylene glycol. BHA and BHT, both of which have one hydroxyl
group, are soluble in fats and oils (up to 50%). Butylated hydroxyanisole (BHA)
and butylated hydroxytoluene (BHT) are antioxidants that are often found in cere-
als, grains, cooking oils, canned goods, snacks, and other similar foods. They pro-
tect foods from oxidative rancidity by contributing hydrogen ions to release fatty
acids [4]. BHA serves as antioxidant and antimicrobial properties. BHA may oper-
ate as a membrane perturbed by inserting into the bacterial cell’s lipid layer and
disrupting the ordered state of the alkyl chains. Because of its alkyl substituents,
BHT may be inefficiently adsorbed at the membrane interface [5]. TBHQ is a
hydroquinone derivative containing a tert-butyl group in the phenolic ring, which
gives this molecule a more hydrophobic property, allowing it to penetrate gram-
positive bacteria .
Natural food colorants are still used around the world and are proven to provide
considerable benefits when taken, are in high demand due to their long or short-term
impacts, as well as their reliability, functionality, biological potential, and health
effects. To be termed natural, food color additives must exist in nature and contain
natural raw components. They can be derived from plants, animals, minerals, or
microorganisms, and the chromophore should not be chemically altered during
extraction or production [6]. Natural colorants are perceived as safer by consumers
than synthetic colorants, which are regarded to be hazardous. These chemical effects
on children can manifest as behavioral problems, hyperactivity, neurotoxicity, and
attention deficiencies, with significant individual variability [5]. In addition to the
detrimental impact of synthetic colorants on health, natural colorants have a favor-
able influence on health. The carotene, a key pigment in skin protection and cell
growth, revealed a beneficial association between health and carotenoids. Lycopene,
a natural pigment present in tomatoes, is helpful in lowering the incidence of all
types of cancer, including breast, prostate, and cervical cancer. Curcumin has been
shown to be a potent antioxidant that protects against oxidative damage in cellular
components and is useful in the treatment of wounds and burns. Few of the natural
color tend to have antioxidant, anti-inflammatory, and pharmacological properties.
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 591
However, due to safety concerns, consumers choose natural colorants over syntheti-
cally generated colorants [7].
Anisochilus carnosus (L) Wall is an annual herb; flowers of this plant are pale
purple, in dense cylindric spikes. They are also known as Karpuravalli and
Padukurkka and are commonly distributed in moist mixed forests in southern hemi-
sphere. These plants have medicinal value because they contain chemically active
compounds that have specific physiological action on the human body. Alkaloids,
flavonoids, tannins, and phenolic compounds are some of the most essential chemi-
cally active constituents of plants [8]. These are multipurpose ethnomedical plants
that are used to treat variety of ailments such as gastrointestinal disorders, respira-
tory problems, cold, cough, and fever. It has also been stated that plant has anti-
inflammatory, anti-ulcer, antifungal, and anticancer properties [9]. Natural colors
originate from sources like plants, animals, insects, and minerals that are found in
nature. Plant-based pigments provide a wide variety of medicinal benefits among all
natural colors [10]. A. carnosus is commonly known as a color yielding plant.
Therefore, the present study was aimed to study the functional groups and antioxi-
dant and antibacterial properties of biocolorant from Anisochilus carnosus for better
understanding the food preservation practice.
2 Results
The extract yield percentage, total phenolics, and flavonoids content of A.carnosus
extract were analyzed, calculated, and results are tabulated in Table 1. The 70%
methanol solvent yielded 1.77% extract recovery under maceration extraction
method. The methanolic extract of A.carnosus showed total phenolics content of
178.98 ± 7.57 mg GAE/g extract and flavonoids were found to be 299.33 ± 14.46 mg
RE/g extract.
The UV-Vis spectroscopy and thin layer chromatography were performed for
qualitative analysis of A.carnosus extract. The spectra of UV-Vis spectroscopy of
methanolic flower extract of A.carnosus were recorded between 200 and 1000 nm
and are presented in Fig. 1. The methanolic extract of A.carnosus showed high
absorbance intensity between ranges of 200–500 nm with a peak absorbance at
263 nm. In TLC analysis, the accumulation of phyto-compounds and pigments in
the extract was higher in the 70% methanol solvent. Both analyses confirmed that
the A.carnosus extract has higher amount of pigments, aromatic phytocompounds,
hydroxyl, carbonyl, and other chromophores groups.
(263.0, 0.7943)
(285.0, 0.7246)
0.9
0.8
0.7
(383.0, 0.4839)
(246.0, 0.6684)
0.6
Abs
0.5
0.4
(351.0, 0.4085)
0.3
0.2
0.1
0.0
200 300 400 500 600 700 800 900 1000
Wavelength(nm)
The FTIR spectral analyses were carried out to understand the functional compo-
nents and also predict the pharmaceuticals groups is present in the A.carnosus
(Tables 2 and 3, Fig. 2). FTIR analysis revealed that A. carnosus methanolic extract
showed beneficial number of phytochemicals and its functional groups which were
responsible for the antioxidant and antibacterial activities. The spectra of A.
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 593
carnosus methanolic extract sample have a different characteristic peak value with
various functional compounds as presented. A broad peak was found around
2930–2850 cm−1 and showed stretching of C–H bond which was attributed to
alkanes group. Stretching of C=O bond around 1730 cm−1 indicated the presence of
594 S. Thamburaj et al.
Fig. 2 FTIR spectrum of A. carnosus extract (a); Chloramphenicol (b); Quercetin and Rutin (c);
BHT and BHA (d)
carboxylic group. The stretching of C=C bond was observed around peak
1690–1660 cm−1 which correspond to alkenes group. The peak around 1370 cm−1
showed medium intermolecular bending vibration of C=O and the peak around
1330 cm−1 showed stretching of C-O bond. The alcoholic group attributed to peak
around 1260–1000 cm−1 which indicated the presence of intermolecular stretching
of C-O bond. The presence of aromatic compounds was observed in a peak around
1460 cm−1 (C=C bending) and 758 cm−1 (C-H bending), respectively.
The major pharmaceuticals groups in A. carnosus methanolic extract were iden-
tified by FTIR spectral analyses with Shimadzu FTIR library (IRs Pharmaceuticals
library). The FTIR spectrum showed the presence of most important pharmaceutical
compounds with similar functional groups in the A. carnosus methanolic extract
(Table 3). Pharmaceutical compounds with their library ID, molecular formula, and
molecular weight (MW) in the extract are given in Table 6. Twenty major pharma-
ceuticals chemical compounds were found in A. carnosus methanolic extract. All
chemical compounds have diversity of pharmaceutical properties and were identi-
fied by these analyses. Similarly, the natural (Quercetin and rutin), synthetic anti-
oxidant (BHA and BHT), and chloramphenicol (Antibacterial agent) functional
groups were compared with A. carnosus methanolic extract under FTIR spectros-
copy analysis. This analysis confirmed that A. carnosus has antioxidant and antibac-
terial functional groups.
The antioxidant activities of free radical scavenging for A.carnosus extract were
studied and results are illustrated in Table 4. The DPPH scavenging activity was
calculated as % inhibition for methanolic extract of A.carnosus which showed that
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 595
Quercetin has higher scavenge activity for DPPH radical and the lowest was
recorded for BHA. A.carnosus extract showed higher antioxidant activity than the
synthetic antioxidants (BHT and BHA) indicating it to be a potential antioxidant. As
per the results, A. carnosus extract had the degree of reduction of 118.94 mM Fe
(II)/1 mg which was greater than BHT (28.29 mM Fe (II)/ 1 mg) and rutin (109 mM
Fe (II)/ 1 mg), whereas A. carnosus extract value was almost equal to BHA
(125.91 mM Fe (II/ 1 mg).When A. carnosus extract was compared to quercetin, it
has shown highest degree of reduction 253.43 mM Fe (II)/1 mg.
The antibacterial activity of methanolic extract of A. carnosus was tested for dif-
ferent pathogens such as S. aureus, S. abony, P. aeruginosa, B. cereus, S. typhi,
E. coli, and S. flexneri (Table 5 and Fig. 3). The antibacterial effect of methanolic
extract of A. carnosus was evaluated using agar well diffusion method. The diame-
ter of ZOI was calculated for each pathogen which includes a well size of 6 mm in
diameter. The higher antibacterial activity was observed against S. flexneri (ZOI
28.16 ± 0.28 mm; MIC 400 μg/mL; MBC 450 μg/mL). The A. carnosus methanolic
extract was tested against the pathogens and was compared with the standard chlor-
amphenicol. The pathogen S. typhi showed higher susceptibility against A. carnosus
methanolic extract with ZOI of 28.00 ± 0.50 mm; MIC 400 μg/mL; MBC 450 μg/
mL; respectively. Similarly, the antimicrobial activity for other pathogens such as
596 S. Thamburaj et al.
Table 6 Total aerobic bacterial count for stored health mix with A. carnosus extract
Incubation period Control (CFU/gm) Treated (CFU/gm) % Inhibition
0 week 2.7 × 105 2.5 × 105 05.18
1 week 6.2 ×105 1.9 × 105 68.48
2 week 1.3 ×106 1.2 × 105 90.39
3 week 2.6 ×106 9.8 × 104 96.23
4 week 6.8 ×106 1.4 × 105 97.80
Control: Untreated health mix with target bacterial cultures; Treated: Health mixes with target
bacterial cultures were thoroughly sprayed with mg/ml A. carnosus extract and left to air-dry
Fig. 3 A. carnosus (a); Color from A. carnosus (b); Antibacterial activity- B. cereus (c) and
S. typhi (d)
S. aureus (ZOI 23.66 ± 0.57 mm; MIC 450 μg/mL; MBC 500 μg/mL), S. abony
(ZOI 23.16 ± 0.76 mm; MIC 450 μg/mL; MBC 500 μg/mL), E. coli (ZOI
20.50 ± 0.50 mm; MIC 500 μg/mL; MBC 550 μg/mL), and S. flexneri (ZOI
26.66 ± 0.28 mm; MIC 400 μg/mL; MBC 450 μg/mL) was also observed. An incon-
sequential antibacterial activity was observed in A. carnosus methanolic extract
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 597
against P. aeruginosa and B. cereus as there was no ZOI observed; it may be attrib-
uted to their phytoconstituent‘s inability to control the pathogens. When compared
with standard chloramphenicol, all the pathogens were inhibited and showed a
higher zone of inhibition.
Total plate count for storage of health mix with A. carnosus extract was analyzed
by standard IS method. The storage study of A. carnosus extract against selected
pathogens in health mix is represented in Table 6. Gram-positive organisms include
S. aureus and B. cereus and gram-negative microbes include S. abony, P. aerugi-
nosa, S. typhi, E. coli, and S. flexneri; their inhibition for about 4 weeks was studied
in the present work. The A. carnosus extract showed maximum inhibition (97.80%)
in fourth week in which health mix had only the microbial count of 1.4 × 105 cfu/g
followed by third and second week where inhibition was observed to be 96.23% and
90.39% with microbial loads of 9.8 × 104 cfu/g and 1.2 × 105 cfu/g, respectively.
3 Methods
3.1 Sample Collection
3.2 Preparation of Extracts
3.3 Total Phenolics
The analysis of total phenol content in A. carnosus extracts was measured spectro-
metrically by using Folin-Ciocalteu assay [12]. About 0.5 mL of the flower extract
was mixed with 2.5 mL of 20% sodium carbonate and incubated at room tempera-
ture for 4 minutes. 0.5 mL of 1 N Folin-Ciocalteu reagent was added and the tube
was further incubated for 40 minutes. The blue absorbance was read at 723 nm in
UV-spectrometer (Shimadzu Japan 1800) against the blank for total phenolics. On
the basis of the standard curve: y = mx + c, the results were in the form of gallic acid
equivalents (mg GAE/g); where x is the absorbance and y is the gallic acid equiva-
lents (mg GAE/g).
3.4 Total Flavonoids
The total flavonoid content in A. carnosus extract was determined using a conven-
tional method [13] with some modifications and represented in the form of rutin as
equivalents per gram of extract (mg RAE/g). To the 300 μL of flower extract, 2 mL
of distilled water was added followed by 150 μL of sodium nitrite. At room tem-
perature, the contents in the test tube were allowed for incubation for 6 minutes.
After the incubation, 150 μL of aluminim chloride was added and subjected to
6-minute incubation. Then 2 mL of 4% NaOH was added and incubated for another
20 min. The absorbance of pink color was read at 510 nm at UV-spectrometer. The
results were based on the calibration curve (y = mx + c) where x is the absorbance
and y is the rutin equivalence (mg/g) value.
1 mg/mL concentration of extract) with a capillary tube, 1.0 cm above from the end
of the silica gel plate. The mobile phases used were hexane (60%): ethyl acetate
(30%): methanol (10%). The spotted TLC plates were kept in solvent-filled cham-
ber. A chromatogram was produced by movement of solute (mobile phase) across a
thin layer of silica (stationary phase). After the chromatogram was generated, the
resolved spots were identified by detection under UV light at both longer and shorter
wavelengths of 365 nm and 254 nm, respectively, which helped to detect the colored
and fluorescence compounds [18].
The DPPH radical scavenging activity of the samples was evaluated by the stan-
dardized method of DPPH radical scavenging activity [14]. 2 mL methanolic DPPH
solution (0.1 mM) was added to the samples and incubated for 20 min at
27 °C. Absorbance of the solution was monitored by using UV VIS spectrophotom-
eter at 517 nm. The mixture of methanol, DPPH and standards rutin, quercetin,
BHA, and BHT served as the positive control [28].
The antioxidant activity of A. carnosus extract was assessed according to the proce-
dure mentioned by Pulido et al. [15] with few modifications. The antioxidant poten-
tial of the A. carnosus extract was estimated to analyze their ability to reduce from
Fe3+ to Fe2+. About 100 μl of extract sample was added to freshly prepared 900 μl of
FRAP solution in 40 mM HCl with 2.5 mL of 20 mM FeCl3·6H2O and 25 mL of
0.3 M acetate buffer with pH 3.6 and was incubated at 37 °C in water bath for
30 min. At the end of incubation period, the absorbances were read at 593 nm in UV
spectrophotometer. Blank was served as FRAP reagent, and instead of sample,
600 S. Thamburaj et al.
methanol was added. The FRAP value was expressed as mM Fe (II) equivalents/ mg
extract.
3.10 Test Pathogens
Pure cultures of Bacillus cereus NCIM-2217, Shigella flexneri NCIM 1457 were
obtained from National Collection of Industrial Microorganisms (NCIM), Pune.
And others, Escherichia coli (MTCC- 443), Salmonella enterica Typhimurium
MTCC-98, Pseudomonas aeruginosa MTCC 1934, Staphylococcus aureus MTCC-
737, and Salmonella enterica ser. Abony MTCC- 3858, were obtained from micro-
bial culture collection and gene bank, IMTECH, Chandigarh. The pure culture was
cultured on a nutrient agar medium.
The modified agar well diffusion method was used to assess the antibacterial activ-
ity of A. carnosus flower extracts against various bacterial pathogens. Using sterile
cotton swabs, bacterial broth cultures were swabbed in MHA media plates. A steril-
ized cork borer (6 mm Hi-Media) was used to create the wells on agar plates. The
plates were aerobically incubated at 37 °C for 18–24 h with diluted samples (100 μL
of 10 mg/mL concentration). Chloramphenicol (5 μg) was considered as positive
and negative control. The zones of inhibition (ZOI) (mm) of each sample were mea-
sured after the necessary time of incubation, and the activity index was determined
as well. The reading was taken for three times in total with the average values
reported. The whole experiment was carried out in vitro conditions [16].
MICs was carried out for the serial extract of A. carnosus in broth dilution method,
for those showed ≥8 mm diameter growth ZOI in well diffusion method. The 5 mg/
mL concentration of extract was serially diluted in 900 μL sterile broth (Bacteria-
MHA broth). All the broth were filled with 100 μL of the culture (1 × 105 CFU/mL)
and one was kept as control for each test organisms; further all the culture tubes
were incubated at 37 °C. After incubation, OD values were taken in spectrophotom-
eter (Shimadzu UV-spectrophotometer, UV 1800) at 570 nm. From OD values, the
percentage of growth inhibition and MIC was calculated against test pathogens. The
MBC was evaluated by subculturing the test dilution used in MIC. The culture
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 601
obtained from the MIC test by centrifugation after incubation period was suspended
in 100 L sterile broth and the suspension was swabbed onto the MHA plates and
allowed to incubate for 24–48 h at 37 °C on to a fresh medium and incubated further
for 24 h. MBC was defined as the concentration of plant extract that fully killed the
organisms [16, 28] .
3.13 Storage Study
Commercially available health mix was collected from local market Thanjavur,
Tamil Nadu, India. The health mix was sterilized by autoclaving at 15 lbs. pressure
(121 °C) for 15 minutes and allowed to cool down. After cooling, the health mix
samples were divided into an experimental group and a control group. Experimental
group consists of sterile health mix sample (25 g) which was first sprayed thor-
oughly with extract solution (2× concentration of MIC/g of sample), allowed to dry
in a sterile container. The desired mixed bacterial culture (3 × 105 CFU/ g of sample)
was mixed well and incubated at room temperature set at 26 °C for 4 weeks. The
control group health mix was not treated with extract and placed for incubation at
room temperature for 4 weeks.
Sample groups were monitored every week to determine shelf-life and to subcul-
ture bacterial spoiling agents. Each group that showed bacterial growth was deter-
mined by standard plate count method. The percentage of bacterial inhibition at
every week was also determined.
3.14 Statistical Analysis
All the experimental data were conducted in triplicates and analyzed statistically.
All data were expressed as mean ± standard deviation (n = 3). All the parameters
were analyzed using factorial by SPSS Inc. 25.0 (2020, IL, USA).
4 Discussion
28]. The DPPH activity was measured through the percentage inhibition of extract
with other antioxidant compounds. According to the results, A. carnosus extract had
the inhibition percentage, whereas natural antioxidant such as rutin and quercetin
also had higher inhibition of 47.20% and 40.39%. When compared with synthetic
antioxidants, the A. carnosus extract arrayed higher inhibition percentage. The
highly enhanced DPPH radical scavenging activity of the extract was observed in
the present study. It may be due to the polar nature of the solvent and the extracting
ability of the phenolics from the A. carnosus extract.
The ferric reducing antioxidant power (FRAP) assay is a common antioxidant
method used to measure the reduction of ferric ion (Fe3+)-ligand complex by anti-
oxidants in an acidic medium to the intensely blue-colored ferrous (Fe2+) complex.
The FRAP assay is performed at an acidic pH 3.6 to maintain iron dispersibility
and, more notably, to boost electron transfer. This elevates the redox potential,
resulting in a shift in the dominant reaction mechanism. Antioxidant activity is mea-
sured as an increase in absorbance at 593 nm, and the responses were recorded as
micromolar Fe2+ equivalents or as a percentage of an antioxidant standard [28, 29].
The ferric reducing power of A. carnosus extract revealed that the presence of phy-
toconstituents in the extract has a strong affinity for ferric ions, and thus redox reac-
tions were extinguished through redox reaction. The reducing power of the bioactive
phytoconstituents present in A. carnosus methanolic extract is associated with high
antioxidant activity. When the antioxidant assay of DPPH and FRAP activity is
compared with each other, it was observed that A. carnosus extract value was higher
than synthetic antioxidants (BHA and BHT), whereas A. carnosus extract value was
equal or lesser than the natural antioxidants such as rutin and quercetin because they
are well-known polyphenolic compounds. Similarly, previous studies have shown
that antioxidant potential of methanolic and aqueous extract of A. carnosus leaf
showed a similar scavenging activity when compared with natural antioxidants such
as rutin and ascorbic acid (Bhagat et al., 2011). From the results above, the high
radical scavenging activity of A. carnosus extract indicates it is highly potent and
can scavenge free radicals in the body.
Quercetin is an aglycone that does not have an attached sugar and it is known to
be the most abundant flavonoid molecules from group of polyphenols [30].
Generally, quercetin can be found in a wide range of foods including apples, berries,
Brassica vegetables, capers, grapes, onions, shallots, tea, and tomatoes, as well as
numerous seeds, nuts, flowers, barks, and leaves [31]. Similarly, rutin is a glycoside
combination of flavanol quercetin and disaccharide rutinose. It is also known as
Vitamin P which is found in a variety of vegetables, fruits, and medicinal herbs,
including asparagus and buckwheat. Additionally, buckwheat is regarded as one of
the best dietary sources of rutin [32]. Previously, it is reported that quercetin and
rutin were proved to have several pharmacological activities including antioxidant,
anticarcinogenic, neuroprotective, cardioprotective, and anti-inflammatory activi-
ties [33–36]. Due to exorbitant rate of natural antioxidants, the synthetic antioxi-
dants have been preferred as a preservative in food industry.
In case of synthetic compounds, BHA and BHT are two of several synthetic
antioxidants that are widely used in a variety of product categories such as food,
604 S. Thamburaj et al.
syndrome, renal impairment, and childhood leukemia. Due to the adverse effects of
antibiotics, the natural substances such as plant extracts are being explored more for
their antibacterial activity [41].
The present study confirmed that the A. carnosus extract has good antibacterial
effect against tested food pathogens except P. aeruginosa and B. cereus. Similarly,
previous studies reported that natural extracts of several plants such as Cleistanthus
collinus, Elaeocarpus tectorius, and rubius fruits have shown zone of inhibition
against several food-borne pathogens [13, 42, 43]. According to the result, antibac-
terial activity may be related to the total polyphenolic content of A. carnosus extract.
Hence, it showed that A. carnosus methanolic extract could inhibit the food borne
pathogens.
B.cereus produces two types of toxins; emetic and diarrhoeal causing vomiting
and diarrhea. S. aureus generally causes infections of skin, bacteremia, urogenital
tract, and central nervous system. Likewise, S. abony, P. aeruginosa, and S. typhi
also cause opportunistic infections. Antibiotic resistance in these food pathogens is
a major problem in food industries that keep on challenging the healthcare sector in
worldwide. The appearance and spread of drug-resistant pathogens have signifi-
cantly threatening the existing antibacterial therapy. Therefore, there is a need to
search for a new source of antibacterial chemical substances to control the multidrug-
resistant pathogens (MDR) in food packaging and preservation. This study con-
firmed that the A. carnosus showed higher antibacterial activity against selected
food pathogens, indicating it should be a potential food preservative to control
unwanted pathogens in any food matrix.
The selected bacterial consortium was used for evaluating the preservative prop-
erties of A. carnosus flower extract. The total plate count method was adopted to
estimate the percentage of inhibition of bacterial growth in the sterile health mix
(Multi grains) with A. carnosus flower extract about 4 weeks. According to best of
our knowledge, this is the first study reporting on A. carnosus leaf extract against
the pathogens in health mix. From this investigation, A. carnosus extract showed
maximum inhibition in fourth week and confirmed that the A. carnosus extract has
antibacterial properties to inhibit the unwanted pathogens in food matrix. This may
be due to the flavonoids and antioxidants present in the A. carnosus extract.
Flavonoids can form complexes with extracellular and soluble proteins, as well as
bacterial cell walls. Lipophilic flavonoids have the potential to damage bacterial
membranes as well [44]. Previous studies have reported the antibacterial activity of
flavonoids against wide variety of human and environmental pathogens [45–47].
Antioxidants in leaf extracts have the ability to break the chain reaction and thus
stops the generation of radical oxygen species (ROS) and helps in decreasing the
oxidation stress which may lead to pathogenesis and inflammation [48]. The activ-
ity of antioxidants against pathogens has been mentioned in previous reports
[49–51].
Antimicrobial activities of medicinal plants and isolated natural antimicrobials
have been linked to variety of processes. These natural compounds influence mac-
romolecular metabolic pathways that have been proved to be targets for antibiotic
intervention. Protein and cell wall production are clearly the targets of the broadest
606 S. Thamburaj et al.
5 Conclusion
compared to natural colors, the synthetic colors are not acquired due to its adverse
effects on human health. Hence, consumers tend to adopt the addition of natural
colors in food. A. carnosus methanolic extract showed significant polyphenolics,
antioxidants, and antibacterial activity against various enteric pathogens.
Furthermore, A. carnosus methanolic extract can act as a potent preservative. Thus,
this natural food color can also be of great commercial importance.
References
1. Oplatowska-Stachowiak M, Elliott CT (2017) Food colors: existing and emerging food safety
concerns. Crit Rev Food Sci Nutr 57:524–548. https://doi.org/10.1080/10408398.2014.889652
2. Jadhav RV, Bhujbal SS (2020) A review on natural food colors. Pharma Resonance 2(2):12–20.
3. Lourenço SC, Moldão-Martins M, Alves VD (2019) Antioxidants of natural plant origins:
from sources to food industry applications. Molecules 24:14–16. https://doi.org/10.3390/
molecules24224132
4. Fung DYC, Taylor S, Kahan J (1977) Effects of butylated hydroxyanisole (BHA) and butyl-
ated hydroxytoluene (BHT) on growth and aflatoxin production of Aspergillus flavus. J Food
Safety 1(1):39–51.
5. Gutiérrez-Larraínzar M, Rúa J, de Arriaga D et al (2013) In vitro assessment of synthetic
phenolic antioxidants for inhibition of foodborne Staphylococcus aureus, Bacillus cereus
and Pseudomonas fluorescens. Food Control 30:393–399. https://doi.org/10.1016/j.
foodcont.2012.07.047
6. Grube M, Muter O, Strikauska S et al (2008) Application of FT-IR spectroscopy for control
of the medium composition during the biodegradation of nitro aromatic compounds. J Ind
Microbiol Biotechnol 35:1545–1549
7. Sezgin A, Ayyıldız S, Sezgin AC (2017) Food additives: colorants. Science within Food:
Up-to-Date Advances on Research and Educational Ideas: 87–94.
8. Kiruthiga N, Sathish Sekar D (2278) Studies on phytochemicals and steroid isolation from
N-hexane extract of Anisochilus carnosus. Int J Adv Biotechnol Res:976–2612
608 S. Thamburaj et al.
9. Reshi NA, Sudarshana MS, Girish HV (2017) In vitro micropropagation of Anisochilus carno-
sus (L) Wall. J Appl Pharm Sci 7:098–102. https://doi.org/10.7324/JAPS.2017.70715
10. Scotter MJ (2011) Emerging and persistent issues with artificial food colours: natural colour
additives as alternatives to synthetic colours in food and drink. Quality Assurance Safety Crops
Foods 3:28–39. https://doi.org/10.1111/j.1757-837X.2010.00087.x
11. Shang HM, Zhou HZ, Li R et al (2017) Extraction optimization and influences of drying meth-
ods on antioxidant activities of polysaccharide from cup plant (Silphium perfoliatum L.). PLoS
One 12:e0183001. https://doi.org/10.1371/JOURNAL.PONE.0183001
12. Biswas AK, Chatli MK, Sahoo J (2012) Antioxidant potential of curry (Murraya koenigii
L.) and mint (Mentha spicata) leaf extracts and their effect on colour and oxidative stability
of raw ground pork meat during refrigeration storage. Food Chem 133:467–472. https://doi.
org/10.1016/j.foodchem.2012.01.073
13. Muniyandi K, George E, Sathyanarayanan S et al (2019) Phenolics, tannins, flavonoids
and anthocyanins contents influenced antioxidant and anticancer activities of Rubus fruits
from Western Ghats, India. Food Sci Human Wellness 8:73–81. https://doi.org/10.1016/j.
fshw.2019.03.005
14. Singh R, Shushni MAM, Belkheir A (2015) Antibacterial and antioxidant activities of Mentha
piperita L. Arab J Chem 8:322–328. https://doi.org/10.1016/j.arabjc.2011.01.019
15. Pulido R, Bravo L, Saura-Calixto F (2000) Antioxidant activity of dietary polyphenols as
determined by a modified ferric reducing/antioxidant power assay. J Agric Food Chem
48:3396–3402. https://doi.org/10.1021/jf9913458
16. Thamburaj S, Ramaraj E, Sethupathy S et al (2020) Antibacterial and antibiofilm activities
of diphyllin against fish pathogens. Microb Pathog 145:104232. https://doi.org/10.1016/j.
micpath.2020.104232
17. Karpagasundari C, Kulothungan S (2014) Analysis of bioactive compounds in Physalis min-
ima leaves using GC MS, HPLC, UV-VIS and FTIR techniques. J pharmacogn phytochem
3:196–201
18. Lydia MA, Thamburaj S, Jagadeesan G, et al (2020) UV/Vis Spectrophotometric
Characterization of the leaf polyphenolics content in Elaeocarpus tectorius and its therapeutic
potential against selected urinary tract infection pathogens. Phytomedicine 169–178.
19. Jain PK, Soni A, Jain P, Bhawsar J (2016) Phytochemical analysis of Mentha spicata plant
extract using UV-VIS, FTIR and GC/MS technique. J Chem Pharm Res 8:1–6. Available
online www.jocpr.com
20. Murugan R, Parimelazhagan T (2014) Comparative evaluation of different extraction methods
for antioxidant and anti-inflammatory properties from Osbeckia parvifolia Arn. - an in vitro
approach. J King Saud University Sci 26:267–275. https://doi.org/10.1016/j.jksus.2013.09.006
21. Thamburaj S, Rajagopal V, Palanivel R, Pugazhendhi S (2022) Effect of different drying treat-
ments on total polyphenolics content and in-vitro biological properties of Ficus benghalensis
fruit: a comparative study. Biocatal Agric Biotechnol 39:102249. https://doi.org/10.1016/j.
bcab.2021.102249
22. Saravanan S, Parimelazhagan T (2014) In vitro antioxidant, antimicrobial and anti-diabetic
properties of polyphenols of Passiflora ligularis Juss. Fruit pulp. Food Sci Human Wellness
3:56–64. https://doi.org/10.1016/j.fshw.2014.05.001
23. Muniyandi K, George E, Mudili V et al (2017) Antioxidant and anticancer activities of
Plectranthus stocksii Hook. f. Leaf and stem extracts. Agri Nat Res 51:63–73. https://doi.
org/10.1016/j.anres.2016.07.007
24. Wulandari L, Retnaningtyas Y, Nuri LH (2016) Analysis of flavonoid in medicinal plant extract
using infrared spectroscopy and Chemometrics. J Analyt Meth Chem 2016:1. https://doi.
org/10.1155/2016/4696803
25. Kumar A, Kumari P, Arun K, et al (2016) Phenolic composition, antioxidant activity and FT-IR
spectroscopic analysis of halophyte Sesuvium portulacastrum L. extract. Int Res J Biological
Sci 5(1):1–13
Biocolorant from Anisochilus carnosus: A Natural Food Preservative 609
26. Kavitha A, Mary Kensa V (2019) Preliminary phytochemical screening and FTIR analysis on
Rivina humilis L. (mixture). Adalyajournal 8(9):208–215
27. Kedare SB, Singh RP (2011) Genesis and development of DPPH method of antioxidant assay.
J Food Sci Technol 48:412–422
28. Manoharan AL, Thamburaj S, Muniyandi K et al (2019) Antioxidant and antimicrobial inves-
tigations of Elaeocarpus tectorius (Lour.) Poir. Fruits against urinary tract infection patho-
gens. Biocatalysis and agricultural. Biotechnology 20:101260. https://doi.org/10.1016/j.
bcab.2019.101260
29. Zhong Y, Shahidi F (2015) Methods for the assessment of antioxidant activity in foods. In:
Handbook of antioxidants for food preservation. Elsevier Inc., pp 287–333
30. Li Y, Yao J, Han C et al (2016) Quercetin, inflammation and immunity. Nutrients 8:167
31. Wiczkowski W, Romaszko J, Bucinski A, et al (2008) Quercetin from shallots (Allium cepa
L. var. aggregatum) is more bioavailable than its glucosides. J Nutrition 138(5):885–888
32. Patel K, Patel DK (2019) The beneficial role of Rutin, a naturally occurring flavonoid in health
promotion and disease prevention: a systematic review and update. In: Bioactive food as
dietary interventions for arthritis and related inflammatory diseases. Elsevier, In, pp 457–479
33. Afanas’ev IB, Ostrakhovitch EA, Mikhal’chik EV et al (2001) Enhancement of antioxidant
and anti-inflammatory activities of bioflavonoid rutin by complexation with transition metals.
Biochem Pharmacol 61(6):677
34. Enogieru AB, Haylett W, Hiss DC et al (2018) Rutin as a potent antioxidant: implica-
tions for neurodegenerative disorders. Oxidative Med Cell Longev 2018:1–17. https://doi.
org/10.1155/2018/6241017
35. Ganeshpurkar A, Saluja AK (2017) The pharmacological potential of Rutin. Saudi Pharmaceut
J 25:149–164
36. Lesjak M, Beara I, Simin N et al (2018) Antioxidant and anti-inflammatory activities of quer-
cetin and its derivatives. J Funct Foods 40:68–75. https://doi.org/10.1016/j.jff.2017.10.047
37. Ousji O, Sleno L (2020) Identification of in vitro metabolites of synthetic phenolic antioxidants
BHT, BHA, and TBHQ by LC-HRMS/MS. Int J Mol Sci 21:1–13. https://doi.org/10.3390/
ijms21249525
38. Thakore KN (2014) Butylated Hydroxyanisole. In: Encyclopedia of toxicology, 3rd edn.
Elsevier, pp 581–582
39. Cannon M, Harford S, Darks J (1990) A comparative study on the inhibitory actions of
chloramphenicol, thiamphenicol and some fluorinated derivatives. J Antimicrob Chemother
26(3):307
40. Wilson DW, Nash P, Singh Buttar H et al (2017) The role of food antioxidants, benefits of
functional foods, and influence of feeding habits on the health of the older person: an overview.
Antioxidants (Basel) 6:81. https://doi.org/10.3390/antiox6040081
41. Londonkar RL, Madire Kattegouga U, Shivsharanappa K, Hanchinalmath JV (2013)
Phytochemical screening and in vitro antimicrobial activity of Typha angustifolia Linn leaves
extract against pathogenic gram negative micro organisms. J Pharm Res 6:280–283. https://
doi.org/10.1016/j.jopr.2013.02.010
42. Elangomathavan R, Suman T, Nancy Beaulah S et al (2015) Comparative analysis of
Cleistanthus collinus aqueous leaf extract and fractions for its antibacterial potential. Int J
Enteric Pathogens 3. https://doi.org/10.17795/ijep22946
43. Dailin DJ, Gomaa S, Enshasy H et al (2019) Mycology project view project Tifus KIT diagnos-
tic view project natural colorant for food: a healthy alternative. Int J Sci Technol Res 8:3161
44. Selvi TA, Chandrasekaran B, Murugan D, Satyan SR (2011) Leaf and Seed extracts of
Bixa orellana L. exert anti-microbial activity against bacterial pathogens. J Appl Pharm Sci
1:116–120
45. Mathesius U (2018) Flavonoid functions in plants and their interactions with other organisms.
Plant 7(2):30
610 S. Thamburaj et al.
46. Wu SC, Yang ZQ, Liu F et al (2019) Antibacterial effect and mode of action of flavonoids from
licorice against methicillin-resistant Staphylococcus aureus. Front Microbiol 10:2489. https://
doi.org/10.3389/fmicb.2019.02489
47. Song M, Liu Y, Li T et al (2021) Plant natural flavonoids against multidrug resistant pathogens.
Adv Sci 8:2100749. https://doi.org/10.1002/advs.202100749
48. Afzal F, Khurshid R, Ashraf M, Gul Kazi A (2014) Reactive oxygen species and antioxidants
in response to pathogens and wounding. In: Oxidative damage to plants: antioxidant networks
and signaling. Elsevier Inc., pp 397–424
49. Sharifzadeh A, Jebeli Javan A, Shokri H et al (2016) Evaluation of antioxidant and antifungal
properties of the traditional plants against foodborne fungal pathogens. Journal de Mycologie
Medicale 26:e11–e17. https://doi.org/10.1016/j.mycmed.2015.11.002
50. Sagbo IJ, Afolayan AJ, Bradley G (2017) Antioxidant, antibacterial and phytochemical proper-
ties of two medicinal plants against the wound infecting bacteria. Asian Pac J Trop Biomed
7:817–825. https://doi.org/10.1016/j.apjtb.2017.08.009
51. Salem MZM, EL-Hefny M, Ali HM et al (2021) Plants-derived bioactives: novel utilization as
antimicrobial, antioxidant and phytoreducing agents for the biosynthesis of metallic nanopar-
ticles. Microb Pathog 158:105107
52. Shalayel MHF, Asaad AM, Qureshi MA, Elhussein AB (2017) Anti-bacterial activity of pep-
permint (Mentha piperita) extracts against some emerging multi-drug resistant human bacte-
rial pathogens. J Herbal Med 7:27–30. https://doi.org/10.1016/j.hermed.2016.08.003
53. Ullah F, Ayaz M, Sadiq A et al (2020) Potential role of plant extracts and phytochemicals
against foodborne pathogens. Appl Sci (Switzerland) 10:10
Therapeutic Properties of Nardostachys
jatamansi and Its Applications
in Post-Chemotherapy-Induced Hair Loss
in Cancer Patients
1 Introduction
Hair loss due to chemotherapeutic agents is one of the most common ectodermal
conflicting effects and is rated as one of the most troubling side effects of cancer
therapy. As per the clinical trials done by Howard (Jack) West, MD; Abdul Rafeh
Naqash, MD, we can infer that not all cancer treatments cause hair loss, and among
which it may be total or partial hair loss and may include some areas that can cover
all over the body. Chemotherapy usually acts by interloping with the more rapidly
proliferating cells of the body, including cancer cells and hair follicles. As we are
aware of the fact that chemotherapy is a common cause of hair loss, some chemo-
therapy agents rigorously cause hair loss that are usually complete ones, while other
chemotherapy agents cause hair loss far more rarely [1]. Damage to hair follicles
following susceptibility to toxic chemotherapeutics can cause notable hair loss,
commonly known as chemotherapy-induced alopecia (CIA) [2].
Chemotherapy drugs are powerful medications that attack rapidly growing can-
cer cells. Unfortunately, these drugs also attack other rapidly growing cells in the
human body, including those in your hair roots. Chemotherapy may cause hair loss
all over the human body, not just on the scalp. Sometimes eyelash, eyebrow, armpit,
pubic, and other body hair also falls out. Some chemotherapy drugs are more likely
than others to cause hair loss, and different doses can cause anything from mere
• The hair follicle (HF) is a complex, dynamically vital, and potent skin appendage
that creates a filamentous hair shaft through rapid keratinocyte proliferation and
terminal differentiation [6].
• Hair follicle is composed of a series of concentric keratinocyte layers and even-
tually functions to produce a central hair shaft. Mature hair follicles in the termi-
nal portion can be split up into
–– A permanent, non-cycling upper section
–– A lower section, which is continuously reassembled during the hair cycle [7].
Therapeutic Properties of Nardostachys jatamansi and Its Applications… 613
• Throughout the life of a human being, the hair follicle undergoes continuous
cycles of enfolding and rebuilding. The hair growth cycle can be classified into
three phases:
1. Growth phase (anagen––lasts an average of 2–6 years)
2. Involution phase (catagen––lasts approximately 1–2 weeks)
3. Rest phase (telogen––lasts for around 5–6 weeks) [8].
• After chemotherapy treatment, the normal hair growth cycle is interrupted, lead-
ing to one of the two distinct pathways, dystrophic anagen or dystrophic catagen.
The dystrophic catagen pathway results in substantial hair loss [9].
• Recovery from hair loss is substantially faster if dystrophic catagen pathway is
induced in the process, with a normal anagen phase that must be initiated after
cessation of chemotherapy.
• Trichoscopy has been demonstrated to be a useful tool for the diagnosis of alo-
pecia through non-invasive analysis of hair shaft formation.
In human skin, hair loss (alopecia) starts approximately 2–4 weeks after the initial
course of chemotherapy, and hair begins to regrow 3–6 months after the end of treat-
ment. The degree of hair loss varies, depends on the specific drug (or drug combina-
tions) used and drug dosages and regimen (such as the number of cycles undertaken).
Drugs with a high potential for inducing alopecia include cisplatin, doxorubicin,
cyclophosphamide, cytosine arabinoside, and etoposide. The metabolic and mitotic
processes in rapidly growing hair follicles are damaged due to anti-cancer drugs
leading to the thinning of hair shaft, which becomes fragile. The disruption of hair
growth and eventual hair loss involves conserved intrafollicular events. This includes
the activation of damage response pathways leading to reduced proliferation and
unusual levels of apoptosis in the hair matrix [10].
3 Trichoscopy
• Flame hairs
• Circle hairs along with a black and white hair
• The hair shaft is depigmented, excluding the base; in the base, the damaged fol-
licular melanocytes recover, resulting in re-pigmentation of the hair follicle
[11, 12].
Pohl-Pinkus constrictions of the hair shaft led to the wave-like pattern of the hair
shaft due to the cyclic nature of chemotherapeutic drugs. When chemotherapy drug
is delivered into the patient’s body, the drug blends with the blood and travels to all
the fast-growing regions (cells). Therefore, the hair matrix cells are damaged by the
cessation of propagation of hair and causing thinning of the hair shaft. On termina-
tion of the chemotherapy treatment, the hair shaft production recommences before
being prevented once more during the next round of chemotherapy. This produces a
hair shaft that has a series of constrictions [13].
• A more analytical response to injury results in exclamation marks, in which frac-
turing of hair appears as a regular sign of chemotherapeutic damage. These frac-
tures develop due to the thinning of the hair shaft, which is a direct result of
declining hair matrix proliferation, which is followed by premature telogen
entry [14].
• Kowalska-Olędzka et al. have reviewed that the black dots represent the most
severe cessation of proliferation with total breakage of the hair shaft [15].
• Circle hairs were often present following prolonged chemotherapy, as per the
observations made after the studies [16].
• Miteva M and Tosti A have reviewed in their study that flame hairs, on the other
hand, result from melanocyte damage rather than a loss of keratinocyte prolifera-
tion. They are caused by ectopic melanin accumulation around the damaged hair
shaft [17].
When the chemotherapy treatment was terminated, the number of hair follicles
in the anagen phase of the hair growth cycle increased extensively; however, these
were repeatedly depigmented due to chemotherapy-induced melanocyte loss. After
one year of the termination of CIA, normal hair shaft production usually resumes,
and it was detected that some hairs are pigmented at the base and white at the tips,
which indicated melanocyte accumulation in the hair bulb [15, 16].
3.1 Apoptotic Pathways
3.2 Proliferation of Keratinocytes
The effect of jatamansi is used for protection from hair loss. Ethanol extract of
jatamansi was investigated for hair growth in albino rats. The hair growth activity
that worked on CIA model was studied by various parameters like hair density,
lymphocyte count, and testosterone level, along with histopathological studies. Hair
growth initiation time was reduced to half on treatment with the extract. The time
required for hair growth was also reduced considerably.
616 P. Azhagu Saravana Babu et al.
4.1 Trichoscopy
S. Inui et al. have concluded in their Clinical and Experimental Dermatology that
trichoscopy is the technique of dermoscopic imaging of the hair and scalp. This is a
novel diagnostic methodology that is both simple and non-invasive. This technique
can be used as a convenient bedside tool for diagnosing common hair and scalp
disorders. Trichoscopy shows small nodules along the shaft. In this technique, the
hairs bend and break. Trichoscopy of pili torti (a rare hair condition characterized
by fragile hair) shows twists of flattened hair shafts along their long axis. In pili
annulati (a genetic trait resembling “ringed hair”) hair shafts with alternating seg-
ments of light and dark color bands are visible [48].
There are different types of hair shaft abnormalities that are observed. It includes
the following:
• Exclamation mark hairs (alopecia areata, trichotillomania, chemotherapy-
induced alopecia) studied by L. Peralta and P. Morais in their study [49].
• Pohl-Pinkus constrictions (alopecia areata, chemotherapy-induced alopecia,
blood loss, malnutrition)
• Comma hairs (tinea capitis)
• Corkscrew hairs (tinea capitis)
• Coiled hairs (trichotillomania)
• Flame hairs (trichotillomania)
• Tulip hairs (trichotillomania, alopecia areata)
Trichoscopy allows differential diagnosis of most genetic hair shaft disor-
ders [50].
4.2 Aid of Trichoscopy
By using the technique of trichoscopy, we can assess the damage to hair follicles,
and it would help us understand different chemotherapeutics, dosages, and treat-
ments’ impacts on hair damage. It also provides valuable information on the effi-
cacy of current or new treatments in preventing this.
It is a cap filled with some frozen material, which may be an ice pack or cold pack
that is to be secured to the head during chemotherapy treatment. When the cap
increases temperature due to scalp heat, it is replaced with another cooling cap to
maintain the cold scalp temperature until treatment is finished [50]. The primary
assumption of the working of those caps is that cooling of the scalp results in
Therapeutic Properties of Nardostachys jatamansi and Its Applications… 619
vasoconstriction, i.e., reducing the flow of blood in the scalp microvasculature, and
therefore reduces drug delivery to prevent the damage of the sensitive hair follicles.
Furthermore, Grevelman EG has studied that the reduction in temperature reduces
active drug uptake into the hair matrix keratinocytes [3, 51].
The primary supposition is that cooling of the scalp results in vasoconstriction (nar-
rowing blood vessels), which reduces the blood flow in the tiny blood vessels of the
scalp and thereby reduces drug delivery and damages the sensitive hair follicles [3,
9]. Furthermore, Hussain O et al. have used in vitro human keratinocyte models to
study the effect of cooling on chemotherapy drug-induced cytotoxicity and have
derived that when the temperature of the scalp reduces, the active drug uptake
reduces into the hair matrix keratinocyte [3, 52].
Schneider MR et al. have examined the drug concentrations within the hair
shafts, or the plucked hair follicles would likely provide an important biomarker to
ascertain whether cooling or other potential therapies could prevent the accumula-
tion of toxic drug concentrations [6]. McGarvey EL et al. have studied that measur-
ing the concentration of the drug and correlating it with the levels of hair loss, as
well as the signs of damage stipulated by trichoscopy, give even greater perception
into the protective perspective of these treatments which is enabling enhancement
of treatments for the greatest efficacy on the prevention of CIA [53].
5 Discussion
of mitotic activity in the hair matrix resulting in a slender and crippled portion of the
hair shaft known as Pohl-Pinkus constriction, which is prone to fracture. All the
shafts break at the same time when the extremely slender portion reaches the sur-
face of the scalp. Chemotherapeutic drugs such as doxorubicin, daunorubicin,
docetaxel, and cyclophosphamide are commonly accompanied by hair loss. Other
drugs inculpated with hair loss are paclitaxel and carboplatin, cyclophosphamide,
doxorubicin and vincristine, vincristine and daunorubicin, cisplatin, carboplatin,
and a combination of cisplatin and 5 FU [7].
6 Conclusion
References
8. Hendrix S, Handjiski B, Peters EM, Paus R (2005) A guide to assessing damage response path-
ways of the hair follicle: lessons from cyclophosphamide-induced alopecia in mice. J Invest
Dermatol 125:42–51
9. Paus R, Haslam IS, Sharov AA, Botchkarev VA (2013) Pathobiology of chemotherapy-induced
hair loss. Lancet Oncol 14:e50–e59
10. Botchkarev VA (2003) Molecular mechanisms of chemotherapy-induced hair loss. J Investing
Dermatol Symp Proc 8(1):72–75. https://doi.org/10.1046/j.1523-1747.2003.12175.x
11. Mill P, Mo R, Fu H et al (2003) Sonic hedgehog-dependent activation of Gli2 is essential for
embryonic hair follicle development. Genes Dev 17:282–294
12. Loomis CA, Joyner AL (2011) Nerve-derived sonic hedgehog defines a niche for hair follicle
stem cells capable of becoming epidermal stem cells. Cell Stem Cell. 8:552–565
13. Bleiker TO, Nicolaou N, Traulsen J, Hutchinson PE (2005) ‘Atrophic telogen effluvium’ from
cytotoxic drugs and a randomized controlled trial to investigate the possible protective effect
of pretreatment with a topical vitamin D analogue in humans. Br J Dermatol 153:103–112
14. Pirmez R, Pineiro-Maceira J, Sodre CT (2013) Exclamation marks and other trichoscopy signs
of chemotherapy-induced alopecia. Australas J Dermatol 54:129–132
15. Kowalska-Olędzka E, Slowinska M, Rakowska A et al (2012) ‘Black dots’ seen under trichos-
copy are not specific for alopecia areata. Clin Exp Dermatol 37:615–619
16. Rossi A, Caterina Fortuna M, Caro G et al (2018) Monitoring chemotherapy-induced alopecia
with trichoscopy [published online ahead of print July 11, 2018]. J Cosmet Dermatol 18:575.
https://doi.org/10.1111/jocd.12687
17. Miteva M, Tosti A (2015) Flame hair. Skin Appendage Disord 1:105–109
18. Cheng H, Hong B, Zhou L et al (2012) Mitomycin C potentiates TRAIL-induced apopto-
sis through p53-independent upregulation of death receptors: evidence for the role of c-Jun
N-terminal kinase activation. Cell Cycle 11:3312–3323
19. Haslam I, Xie G, Zhou G et al (2015) Shh signaling regulates the damage response of murine
and human hair follicles in chemotherapy-induced alopecia. J Invest Dermatol 135:S7
20. Adapted alf Paus, Haslam IS, Sharov AA, Botchkarev VA (2013) Pathobiology of
chemotherapy-induced hair loss. The Lancet Oncology 14(2):e50–e59
21. Hughes BT, Sidorova J, Swanger J, Monnat RJ Jr, Clurman BE (2013) Proc Natl Acad Sci U S
A 110(22):8954–8959. https://doi.org/10.1073/pnas.1302927110. Epub 2013 May 13
22. Trueb RM (2009) Chemotherapy-induced alopecia. Semin Cutan Med Surg 28:11–14
23. Jimenez JJ, Yunis AA (1992) Protection from chemotherapy-induced alopecia by
1,25-dihydroxyvitamin D3. Cancer Res 52:5123–5125
24. Hidalgo M, Rinaldi D, Medina G et al (1999) A phase I trial of topical topitriol (calcitriol,
1,25-dihydroxyvitamin D3) to prevent chemotherapy-induced alopecia. Anti-Cancer Drugs
10:393–395
25. Bleiker TO, Nicolaou N, Traulsen J et al (2005) ‘Atrophic telogen effluvium’ from cytotoxic
drugs and a randomized controlled trial to investigate the possible protective effect of pretreat-
ment with a topical vitamin D analogue in humans. Br J Dermatol 153:103–112
26. Lotfi-Jam K, Carey M, Jefford M, Schofield P, Charleson C, Aranda S (2008) Nonpharmacologic
strategies for managing common chemotherapy adverse effects: a systematic review. J Clin
Oncol 26:5618–5629
27. Cline BW (1984) Prevention of chemotherapy-induced alopecia: a review of the literature.
Cancer Nurs 7:221–228
28. Hussein AM, Jimenez JJ, McCall CA, Yunis AA (1990) Science. 249(4976):1564–1546.
https://doi.org/10.1126/science.2218498. PMID: 2218498
29. Hussein AM (1995) Int J Dermatol. 34(7):470–3. https://doi.org/10.1111/j.1365-4362.1995.
tb00612.x. PMID: 7591409
30. Hussein AM (1993) South Med J. 86(5):489–96. https://doi.org/10.1097/
00007611-199305000-00001.PMID: 8488392
31. Imanishi H, Tsuruta D, Tateishi C, Sugawara K, Paus R, Tsuji T et al (2010) Laminin-511,
inducer of hair growth, is down-regulated and its suppressor in hair growth, laminin-332 up-
regulated in chemotherapy-induced alopecia. J Dermatol Sci 58:43–54
622 P. Azhagu Saravana Babu et al.
32. You J, Gao F, Tang H, Peng F, Jia L, Huang K, Chow K, Zhao J, Liu H, Lin Y, Chen J (2019)
A medicinal and edible formula YH0618 ameliorates the toxicity induced by doxorubicin via
regulating the expression of Bax/Bcl-2 and FOXO4. J Cancer 10(16):3665–3677
33. You JS et al (2016) Effect of YH0618 soup on chemotherapy-induced toxicity in patients with
cancer who have completed chemotherapy: study protocol for a randomized controlled trial.
Trials 17(1):354
34. Chan YC et al (2009) Nanonized black soybean enhances immune response in senescence-
accelerated mice. Int J Nanomedicine 4:27–35
35. Yim JH et al (2009) Antinociceptive and anti-inflammatory effects of ethanolic extracts of
Glycine max (L.) Merr and Rhynchosia nulubilis seeds. Int J Mol Sci 10(11):4742–4753
36. Kusano S, Abe H, Tamura H (2001) Isolation of antidiabetic components from white-skinned
sweet potato (Ipomoea batatas L.). Biosci Biotechnol Biochem 65(1):109–114
37. Macduff C, Mackenzie T, Hutcheon A et al (2003) The effectiveness of scalp cooling in pre-
venting alopecia for patients receiving epirubicin and docetaxel. Eur J Cancer Care 12:154–161
38. Mols F, Van Den Hurk CJ, Vingerhoets AJJM et al (2009) Scalp cooling to prevent
chemotherapy-induced hair loss: practical and clinical considerations. Support Care Cancer
17:181–189
39. Kennedy M, Packard R, Grant M et al (1983) The effects of using chemocap on occurrence of
chemotherapy-induced alopecia. Oncol Nurs Forum 10:19–24
40. Lovejoy NC (1979) Preventing hair loss during adriamycin therapy. Cancer Nurs 2:117–121
41. Rodriguez R, Machiavelli M, Leone B et al (1994) Minoxidil (mx) as a prophylaxis of
doxorubicin-induced alopecia. Ann Oncol 5:769–770
42. Granai CO, Frederickson H, Gajewski W et al (1991) The use of minoxidil to attempt to
prevent alopecia during chemotherapy for gynecologic malignancies. Eur J Gynaecol Oncol
12:129–132
43. Villani C, Inghirami P, Pietrangeli D (1986) Prevention by hypothermic cap of antiblastic
induced-alopecia. Eur J Gynaecol Oncol 7:15–17
44. Edelstyn GA, MacDonald M, MacRae KD (1977) Doxorubicin-induced hair loss and possible
modification by scalp cooling. Lancet 2:253–254
45. Peck HJ, Mitchell H, Stewart AL (2000) Evaluating the efficacy of scalp cooling using the
penguin cold cap system to reduce alopecia in patients undergoing chemotherapy for breast
cancer. Eur J Oncol Nurs 4:246–248
46. Jadhav VM, Throat RM, Kadam VJ, Kamble SS (2009) Herbal Anxiolyte, Nardostachs jata-
mansi. J Pharm Res 2(8):1208–1211
47. Yeap SK, Rahman MBA, Alitheen NB, Ho WY et al (2011) Evaluation of immunomodulatory
effect, selection of the correct targets for Immunostimulation study. Am J Immunol 7(2):17–23
48. Inui S, Nakajima T, Itami S (2010) Coudability hairs: a revisited sign of alopecia areata
assessed by trichoscopy. Clin Exp Dermatol 35(4):361–365
49. Peralta L, Morais P (2012) Photoletter to the editor: the friar tuck sign in trichotillomania. J
Dermatol Case Rep 6(2):63–64
50. Wu MY, Li J. (2021) CMAJ. 193(4):E126. https://doi.org/10.1503/cmaj.200894
51. Grevelman EG, Breed WP (2005) Prevention of chemotherapy-induced hair loss by scalp cool-
ing. Ann Oncol 16:352–358
52. Al-Tameemi W, Dunnill C, Hussain O et al (2014) Use of in vitro human keratinocyte models
to study the effect of cooling on chemotherapy drug-induced cytotoxicity. Toxicol In Vitro
28:1366–1376
53. McGarvey EL, Baum LD, Pinkerton RC, Rogers LM (2001) Psychological sequelae and alo-
pecia among women with cancer. Cancer Pract 9:283–289
54. Botchkarev V, Sharov AA, Syska W, Maurer M, Gilchrest BA (2002) Involvement of p53 and
Fas in hair follicle apoptosis in human skin/SCID model for chemotherapy-induced hair loss.
J Invest Dermatol 119:286
55. Food and H.H.S. Drug Administration (2016) Medical devices; general and plastic surgery
devices; classification of the scalp cooling system to reduce the likelihood of chemotherapy-
induced alopecia. Final order. Fed Regist 81(29):7452–7454
Phitotherapeutic Potential of Cassava
(Manihot esculenta, Crantz)
1 Introduction
Cassava as Food
Cassava is the best known roots crop used as food. Results of the evaluation of cul-
tivars selected among the most used in commercial crops in the southeastern region
of Brazil point to a variation in productivity from 25.0 to 36.0 t ha−1, with humidity
around 65% [1]. The percentage of dry mass reflects the starch content, which
approaches 40% (Table 1); this explains more than 1500 kcal kg−1 of raw roots. The
same table highlights the reduced content of other components, such as protein and
lipids, but highlights the presence of dietary fiber, which is close to the recom-
mended amount of 2 g day−1, in whatever form it is consumed, cooked or fried.
Even so, it is necessary to acknowledge that it is a caloric food.
Cassava leaves are not widely known as food, although in Brazil they are con-
sumed in the north and some northeastern regions as a typical dish known as
maniçoba. But fresh leaves can be collected separately from the roots, with mois-
ture around 77% [1], and consumed as a vegetable. Values obtained in Paraná, a
Brazilian state in the southeastern region, allowed us to estimate the production of
leaves at 3400 kg ha−1 year−1, considering the average annual harvest after pruning,
carried out for 3 years for four varieties of commercial use [2]. In this case, the
leaves complement the harvest of the roots as a source of protein, but also of fiber,
cyanogenic, and phenolic compounds. Despite this potential, the international con-
sumption of cassava has been restricted due to the presence of cyanogenic com-
pounds, which are not included in the Brazilian Food Composition Table [3]. This
omission is explained by the lack of dissemination of national and international
research results, which scares consumers who are not familiar with their
M. P. Cereda (*)
University of São Paulo State (UNESP), Botucatu, São Paulo, Brazil
Table 1 Cassava root composition in raw, cooked and fried, according to the (Brazilian Food
Composition Table [3]) and literature data
Cassava Leaves
Food products Unity Crude Cooked Fried Fresh Dehydrated (**)
Energia (kcal 100 grams−1) 151 125 300 -*- 469.3
Moisture (g 100 grams−1) 61.8 68.7 38.8 (**) (**)
Ash (g 100 grams−1) 0.6 0.4 0.6 0.70 7.37
Fat (g 100 grams−1) 0.3 0.3 11.2 0.70 7.36
Protein (g 100 grams−1) 1.1 0.6 1.4 3.48 22.36
Carbohydrate (g 100 grams−1) 38.2 30.1 50.3 3.90 10.84
Dietary fiber (g 100 grams−1) 1.9 1.8 1.9 1.21 (*) 52.07 (*)
Note: part of the nitrogen present, used to establish the protein content, may come from the CN-
radical (FAO/WHO [13])
-*- data not available, (*) as total fiber, (**) data from Fasuyi [71], adjusted for dry basis
consumption and makes exporting cassava difficult. It is common for cassava flour
to be exported as “animal feed” for consumption by expatriate Brazilians.
To justify the potential of cyanogenic compounds, the review by Cressey and
Reeve [4] and others, for example, for human and animal poisoning by Gensa [5],
for phenolic compounds, a review of functional fibers [6], and as a phitotherapic by
Fuentes-Zaragoza et al. [7], was used as a basis for this chapter. Especially with
respect to the toxic effects of cyanide, justifications will be presented as to why the
risk of its consumption is of little concern if the consumer is well nourished.
It has long been known that some cultivated plants have toxic principles, but only
around the year 1700 was cyanide recognized for its pharmacological properties,
even though it is a toxic substance widely present in food crops. In 1817, Magendie
suggested that Hydrogen cyanide (HCN), in appropriate doses, could be used to
control cough and lung diseases. This assumption was also accepted by Granville,
an English physician, who included HCN-rich wild cherry water in the British
Pharmacopoeia, where it remained until 1948 [8].
Seigler [9] pointed out that cyanogenic glycosides occur in more than 2650 plant
species, from 550 genera and 130 families. These plants are responsible for the
production of more than 60 cyanogenic glycosides, some of which are present in
edible plants. Among the plant species that produce cyanogenic glycosides, many
also produce a hydrolytic enzyme from the β-glycosidase group. The enzyme and
cyanogenic glycosides remain in separate compartments of the plant [10] and only
come into contact when this cellular structure is disrupted, with subsequent release
of HCN, in addition to an aldehyde or a ketone [11, 12]. The release of HCN from
cyanogenic glycosides can also result from enzymatic hydrolysis by intestinal
microflora [13, 14]. Among the plants that present glycosides, cassava stands out for
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 625
its status as a staple food in tropical regions. Nambisan [15] reports on its consump-
tion in connection with food security and its economic importance, since it is tradi-
tionally used as a reserve in case of natural disasters, such as cyclones and droughts.
Cassava produces reasonable yields in low-fertility soil with a flexible harvesting
period, which makes it a crop of great importance in many countries, but always
with a focus on subsistence food. According to Onyenwoke and Simonyan [16] the
postharvest activities with cassava include grating or grinding, followed by drying,
operations that are not complicated and do not require large investments, so they can
be carried out in rural areas, on farms or in farming villages. In addition to its con-
sumption as a subsistence food, cassava roots have increasingly been traded interna-
tionally, in part because of the migration of consumers from countries where it is a
staple of the diet to other parts of the world where there is no tradition of consump-
tion. Knudsen et al. [17] point out that in cooking, cassava is consumed in various
ways, such as flour, chips, grated root mass (baked, steamed, or fried), whole
steamed roots, and tapioca pearls.
Considerable confusion exists concerning the forms used in the literature, standards
and legislation that deal with cassava cyanogenic glycosides and their metabolites,
because, unlike other toxic substances, nontoxic organic molecules bind to a radical
toxin to all forms of life. For more in-depth knowledge, the reader is encouraged to
read the reviews of Cressey and Reeve [4] on the metabolism of cyanogenic glyco-
sides and Rosling [18] on cassava toxicity and food security. These texts will deepen
the reader’s knowledge about the presence of cyanogenic compounds in cassava.
The term “HCN” is recommended by the International Union of Pure and
Applied Chemists (IUPAC) [19] to indicate hydrogen cyanide, but it is not necessar-
ily appropriate when the cyanide radical comes from a cyanogenic glycoside. These
forms of expression and spelling cause considerable confusion, so this chapter will
follow the recommendations of Bokanga et al. [20], who prefer and justify express-
ing the cyanogenic potential of a food (CN−p), which is spelled differently, in the
form “total hydrocyanic acid” or “bound” or “free and combined” hydrocyanic acid.
The free cyanide radical may be denoted by CN−. Because it is clear and precise,
this concept and spelling have been adopted in this chapter.
According to Conn [10], cyanide is a common substance in the environment and
can affect both humans and animals [5]. According to Rosling [18], once released,
the CN− radical can be solubilized in water to form HCN, establish secondary cya-
nide compounds, or simply volatilize.
Vegetables are an important source of cyanide, present in the form of cyanogenic
glycosides. Table 2 highlights the most common cyanogenic glycoside-containing
food crops and their botanical sources. Amygdalin and prunasin are the best-known
cyanogenic glycosides and are present in the seeds of apricots, apples, and other
plants. Linamarin and lotaustralin, in addition to other sources, are both present in
626 M. P. Cereda
cassava (Manihot sp.). In all examples, the CN− radical is linked to glucose by a
ß-type bond that can only be broken by the action of a β-glucosidase enzyme, releas-
ing cyanide.
Ingestion can release cyanide in the intestine, where the environment is alkaline,
favoring the action of linamarase [18], human β-glucosidase [21], or bacterial
enzymes, which results in the release and absorption of cyanide in the small
intestine.
Among the plants listed in Table 2, cassava stands out for being one of the few spe-
cies in which the cyanogenic glycoside is found in edible parts of the plant (roots
and leaves). The main toxic principle is linamarin [22], which coexists with its
methyl homologue, called methyl linamarin or lotaustralin (Fig. 1), both of which
occur in variable amounts in all parts of the plant, with a predominance of linama-
rin, over 90%.
The synthesis of linamarin and lotoaustralin occurs in the leaves and roots of
cassava throughout all its botanical cycle [23], but the leaves have the highest con-
tent of both glycosides and the enzyme linamarase [24]. In cassava, lotoautralin
occurs at low concentrations, which is why this chapter will only use linamarin as a
reference.
The literature reports that in cassava, potential HCN concentrations can vary
greatly with altitude, geographic location, and seasonal and production conditions
[25]. Cardoso et al. [26] considered cassava roots as sweet or bitter based on the
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 627
Fig. 1 Structural formulas of Linamarin e Lotaustralin. (Source: Behera and Ray [101]. Published
in CRC Press)
Fig. 2 Summarizes the release of HCN from Linamarin. (Source: Adapted from Sayre et al. [102]
and Gomez and Stuefer [103])
Table 3 Distribution of total potential cyanide by cassava root parts selected for food use
Potencial Cyanide
Portion of the root Weight Moisture (CNp)
% % mg/kg %
Root Entire 100.00 57.0 32.18 100.0
Córtex Inner bark 14.0 76.0 156.16 81.0
Reserve parenchyma Pulp 85.0 58.0 6.12 19.0
Source: Cereda [1]
Cressey and Reeve [4] reviewed the literature on the metabolism of cyanogenic
glycosides, which is the key to the safe consumption of foods that may contain these
compounds. The authors point out that the potential for toxicity of cyanogenic gly-
cosides arises from enzymatic degradation, which produces hydrogen cyanide.
Information on the metabolism of cyanogenic glycosides is available from in vitro,
animal, and human studies. In the absence of β-glucosidase enzymes from the
source plant material, two processes appear to contribute to the production of
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 629
cyanide from cyanogenic glycosides. The first one is the proportion of the glycoside
dose that reaches the large intestine, where most of the bacterial hydrolysis occurs,
and the rate of hydrolysis of cyanogenic glycosides to cyanohydrin and cyanide.
Some cyanogenic glycosides, such as prunasin, are actively absorbed in the jejunum
by utilizing the epithelial sodium-dependent monosaccharide transporter (SGLT1).
The rate of cyanide production from cyanogenic glycosides due to bacterial
β-glycosidase activity depends on the sugar moiety in the molecule and the stability
of the intermediate cyanohydrin following hydrolysis by bacterial β-glucosidase.
Cyanogenic glycosides with a gentiobiose sugar, amygdalin, linustatin, and neoli-
nustatin, undergo two-stage hydrolysis, with gentiobiose initially being hydrolyzed
to glucose to form prunasin, linamarin, and lotaustralin, respectively. While the
overall impact of these metabolic factors is difficult to predict, the toxicity of cya-
nogenic glycosides will be less than the toxicity suggested by their theoretical
hydrocyanic acid equivalents.
It is accepted that the potential toxicity of a food produced from a cyanogenic
plant also depends on reducing, during preparation, the concentration of HCN to
harmless levels for human exposure after consumption [38]. In order for this reduc-
tion in toxicity to occur and for the residual cyanide content to remain at a safe level,
the amount of active available ß-glucosidase must be adequate, in addition to having
temperature and pH conditions for the reaction to occur, neither of which is always
well established.
According to Logsdon et al. [39], three main factors determine the degree of
toxicity of HCN: amount to which the organism is exposed, exposure route, and
exposure time. Therefore, it is clear that it is the released CN− radical that causes
damage to health, whether due to acute intoxication or the risks of prolonged con-
sumption. To express full CN toxicity, several factors must be considered. Closed
environments are more prone to poisoning symptoms than open, ventilated environ-
ments, but once inhaled, the risks are severe.
However, the release of cyanide as a gas allows the formation of stable com-
plexes with metals with which CN− has a high affinity. Especially with iron, the
reaction forms potassium ferrocyanide (K4[Fe (CN)6]), where each iron molecule
can immobilize six CN− molecules and give rise to a blue complex (Prussian blue)
[40]. In water, CN− disperses spontaneously as HCN, but when the equipment used
to extract starch or process cassava roots is made of iron, it is common for the prod-
ucts to show blue spots.
The FAO/WHO [41] reports that cyanogenic glycosides can cause acute poison-
ing in humans, as well as several chronic diseases associated with the consumption
of underprocessed cassava.
In relation to human beings, the most serious problem of toxicity occurs in inha-
lation. In this case, the CN− radical can easily bind iron in the ferric state (Fe3+) of
mitochondrial cytochrome oxidase, in the enzyme that mediates the transfer of elec-
trons to molecular oxygen, the last step of oxidative phosphorylation. This affinity
allows free cyanide to combine with hemoglobin to form cyanohemoglobin, block-
ing electron transfer between cytochrome b and cytochrome c1. As a consequence,
ATP formation is inhibited [42, 43]. The mechanism of mitochondrial oxidation
630 M. P. Cereda
inhibition interrupts the electron transport chain of the inner mitochondrial mem-
brane [44] and, consequently, the transport of oxygen [42], producing tissue anoxia.
The reduction in tissue oxygen utilization produces a state of histotoxic anoxia.
Cyanide bound to the ferric ion of cytochrome c oxidase reduces by 90% the total
uptake of oxygen in most cells through the electronic transport chain [45]. Clinical
symptoms associated with acute toxicity may occur within a few minutes with head-
ache, nausea, vomiting, dizziness, palpitations, hyperpnea and then dyspnea, brady-
cardia, unconsciousness, and violent convulsions, followed by death, depending on
the dose of CN− [38]. Cyanide is not cumulative, so either death from asphyxia and
cardiac arrest occurs or the body is able to eliminate the cyanide in urine, in the form
of ammonium thiocyanate, which is also used as a reference for cyanide intoxication.
If, when inhaled in the form of HCN, cyanide gas is rapidly absorbed from the
respiratory tract, absorption from the gastrointestinal tract is slower, even when cya-
nide salts (sodium and potassium salts) and cyanogenic glycosides are ingested.
Poisoning by skin contact with concentrated solutions of sodium and potassium
cyanide may also occur, with absorption being even slower than by inhalation [46].
When consumed and absorbed in the intestine, linamarin is transported in the
blood, always in the form of an intact glycoside. According to Philbrick [37], it will
be excreted unchanged in the urine, without causing any harm to the organism.
If the lethal dose is reached or exceeded, there is a risk of death, but the lethal
dose is related to the body weight of the consumer.
Lethal Dose
Burtis and Ashwood [47] remind us that the limit between intoxication and death by
a toxic substance is called the lethal dose (LD), which is established in experiments
with animals, never using humans as test subjects. The LD represents the amount of
toxic substance that in a single dose causes the death of all animals, while LD50 is
the single dose that causes the death of 50% of the animals used in the test. The LD
for linamarin from cassava cited by Joint FAO/WHO food standards programme is
10 mg HCN kg−1 body weight [13].
To establish the LD of a compound, it is very important to establish how it is
consumed. For cassava and its derivatives, the culinary products are ingested and
not inhaled. Therefore, it is more appropriate to express the results in potential cya-
nide (CN−p), as recommended by Bokanga et al. [20]. Many of the experiments to
determine the LD were set up with HCN or with potassium cyanide, an inorganic
compound where CN− is covalently linked. That is why it is important to establish
the LD with the cyanogenic compound present in cassava roots, in this case
linamarin.
To establish this limit, Ramalho et al. [32] used linamarin extracted from cassava
cultivars and administered orally to mice and obtained a LD50 of 35.35 mg of CN ±
1.5 mg kg−1 of body weight seven times higher than the LD established by FAO/
WHO. The surviving animals did not show anoxia in noble tissues (cortex neurons
and Purkinje cells), nor were significant changes detected in the biochemical levels
of albumin, glucose, alanine transaminase, aspartate transaminase, and lactate dehy-
drogenase. The results obtained by the authors would allow us to predict the LD for
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 631
According to Esser [48] and Oke [49], if the limits for acute intoxication are not
reached, the cyanogenic compounds in cassava can be eliminated by a detoxifica-
tion mechanism, which involves a two-step reaction. The first step involves the
binding of cyanide to a protein. In the second step, cyanide binds to heme iron.
The first step in the detoxification mechanism occurs with the reaction of cyanide
with sulfur (S) to form thiocyanate, which may have the enzyme rhodanase as a
catalyst. Thiocyanate is a less toxic compound that is eliminated via the kidneys
[46]. Diniz et al. [50] pointed out that the source of sulfur could be cysteine, a sulfur
donor amino acid, to give rise to thiocyanate, which is eliminated in the urine. Since
the effect of linamarin, acetone cyanohydrin, and/or HCN is not cumulative, sub-
lethal doses depend only on available S for cyanide to be slowly and efficiently
eliminated from the body, which depends on adequate nutrition. Piste [51] pointed
out that meat is a good source of cysteine.
Therefore, a balanced diet that contains meat is a decisive factor in circumvent-
ing the deleterious effect of the presence of cyanide residues. Chronic conditions
after prolonged exposure to low levels of cyanide can result in breathing difficulties,
eye irritation, chest pain, vomiting, loss of appetite, headaches, nosebleeds, an
enlarged thyroid gland (goiter), and death. Tropical ataxic neuropathy (TAN) is also
a syndrome attributed to exposure to cyanide from improperly prepared cassava.
Other diseases or syndromes related to the consumption of cassava food and prepa-
ration processes could be mentioned, but the relevant reports, such as in the case of
Konzo syndrome, are still poorly substantiated.
Kashala-Abotnes et al. [52] highlight that Konzo is an irreversible upper motor
neuron disease in sub-Saharan Africa, generally linked to the chronic consumption
of insufficiently processed cassava. Epidemics of neurodegenerative diseases puta-
tively caused by food toxins have been reported in the tropics with no clear under-
standing of their pathogenetic mechanisms. Factors of susceptibility, including
genetics, poor nutrition, poverty, and dietary cyanide exposure, or their interactions,
have been proposed. For the WHO [53], the consequences of chronic exposure to
subacute toxic doses of CN− may be involved in the pathogenesis of certain condi-
tions, including disturbances of thyroid function in the presence of iodine deficiency
and neuropathies. Human populations that eat cassava have ophthalmic and neuro-
logical symptoms associated with exposure to CN−, although it is likely that other
nutritional or metabolic deficiencies that affect the cyanide detoxification
632 M. P. Cereda
differentiate seven groups, ranging from 2.57 to 24.35 mg CN−p kg−1 [56]. These
values are much lower than the range cited by Sulyok et al. [57] in flour samples
collected in Tanzania, with values ranging from 0.45 to 50.0 mg CN−p kg−1 with a
sample with 400 mg of CN−p kg−1.
The example analyses carried out in Africa to confirm the limits established by
the Coordinating Committee for Africa (CCAFRICA). FAO/WHO [58–60] results
of HCN dosage in processed foods in Nigeria showed that the incidence of HCN in
fresh cassava samples was 6 in 8, while in dried cassava all 12 samples showed the
presence of HCN. In another study considering 10 samples of fermented cassava
flour called gari, it was possible to detect HCN above 1.5 mg CN−p kg−1 in half of
the samples, but HCN was not detected in 10 samples of fufu. This traditional food
is made from whole or pieces of roots that are naturally fermented for 3–7 days. At
the same time, 9 out of 10 samples of cassava flour without fermentation had HCN,
with 5 above 2.0 mg of HCN kg−1. In summary HCN was detected at high rates in
fermented cassava products, but in low concentrations. Experts say that processing
can reduce HCN to a minimum or eliminate it.
These results are explained when the conditions for the action of linamarase are
analyzed, where the local temperature was probably within the optimal range of
30–45 °C for the activity of the enzyme. On the other hand, fresh samples probably
had higher water activity, so the enzyme had a greater opportunity to remain active
and hydrolyze linamarin, releasing HCN to the atmosphere, which did not occur in
the dry samples, where the low humidity kept the enzyme inactive, generating high
levels of inactive CN−p. As for the results for fermented cassava products, the pH of
fresh cassava is within a range of 5.0–6.0, but cassava fermentation favors the pres-
ence of lactic acid bacteria, which reduce the pH to values below the optimum.
Paralyzing the action of the enzyme. Reporting of results obtained for ready-to-eat
cassava chips marketed in Australia and New Zealand showed levels equivalent to a
limit of 10 mg HCN kg−1 body weight. These chips are obtained by frying at tem-
peratures above 120 °C, which certainly inactivates the enzyme responsible for
releasing cyanide.
The fact that the cyanide content present in cassava roots is influenced by several
factors, including climatic conditions and cultivation, limits the only factor that can
be considered safe for cassava-derived foods, the determination of the potential cya-
nide content (CN−p) and free cyanide (CN−), keeping in mind that the potential
cyanide content can transform into free cyanide if conditions are favorable for the
occurrence of hydrolysis, enzymatic or acidic, even in the human body, during
digestion.
Significant cases of cyanide poisoning are reported only among people whose
diet is composed almost exclusively of cassava and, in addition, are malnourished.
Compared with African countries, there are few reliable reports of cyanide poison-
ing in South America, where consumption can be high, but malnutrition is rarer.
634 M. P. Cereda
As already mentioned, the cassava plant is more analyzed and referred to as a sub-
sistence food. There are few references to phenolic compounds.
According to Rickard [63] and Padmaja [64] the phenolic compounds that pre-
dominate in the cassava plant are tannins. Ferraro et al. [65] discuss cassava root as
an example of a variety of phenolic compounds, and Uarrota et al. [66] cite ascor-
bate and guaiacol peroxidase as enzymes with antioxidant effects.
Yi et al. [67] report on the occurrence of 10 phenolic compounds in cassava stalk
grown in Hainan in southern China. The compounds were extracted with ethyl ace-
tate and n-butanol and were identified as coniferaldehyde, isovanillin,
6-Deoxyisojacareubin, scopoletin, syringaldehyde, pinoresinol, p-Coumaric acid,
ficusol, balanophonin, and ethamivan, all with significant antioxidant activities. The
results of the study showed that these phenolic compounds contributed to the anti-
oxidant activity of cassava.
In the same material, Suresh and Saravanakumar [68] reported that stalk was a
waste product in cassava root harvesting. It is a renewable raw material rich in anti-
oxidant compounds. To take advantage of them, the authors developed a procedure
to extract anthocyanins and analyze their antioxidant properties. Total phenol, quer-
cetin, and gallic acid were measured. The authors concluded that the phenolic com-
pounds and anthocyanins were responsible for the free radical and that the cassava
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 635
leaf stalk could be used as a valuable source of anthocyanin extraction that would be
applicable in clinical settings and in the food industry.
Cassava leaves are the organs with the most reported phenolic compounds in the
literature. Some authors discuss the collection of leaves specifically for human or
animal consumption. Ravindran and Ravindran [69] evaluated their use for animal
feed, while Ortega-Flores et al. [70] used leaves from shoot pruning focused on
human consumption, evaluating drying in an oven with air circulation and lyophili-
zation. Table 4 shows the levels of phenolic compounds in cassava leaves dehy-
drated by heat in an oven (50 °C) and by lyophilization. The results show that in
relation to lyophilization, which better preserves phenolics, the leaves maintained
high levels of potential cyanide content.
Ravindran and Ravindran [69] established the variation of nutritional value at
three stages of cassava leaf maturity (very young, young, and mature leaves). The
nutritional composition and antinutritive properties of the crude protein and carbo-
hydrate contents decreased with maturity, whereas all other proximal and fiber com-
ponents increased. The mineral profile showed cassava leaves to be good sources of
most minerals, particularly of calcium and trace minerals. The phosphorous and
sodium contents, however, were low. The values for K, Mg, P, Zn, and Mn decreased
with leaf maturity, while those for Ca, Na, and Fe increased. The researchers also
found that cassava leaves were rich in all essential amino acids except methionine
and phenylalanine. As the leaves matured, the tendency was for the amino acid con-
centrations to decrease. Only glutamic acid, proline, and glycine contents increased,
while those of valine and phenylalanine were unaffected. The levels of phytic acid
increased with leaf aging, while tannin and hydrocyanic acid contents decreased.
Fasuyi [71] worked in animal feed aiming to reduce the content of cyanide and
phenolics in it, which are considered as antinutritional compounds. The HCN con-
tent in fresh leaves varied from 40 to 60 mg 100 g−1, phytates ranged from 107 to
249 mg 100 g−1, and tannic acid varied from 7.5 to 15.0 mg 100 g−1. The author
evaluated the cassava leaves of four Pakistani varieties, researching drying pro-
cesses, sun-drying, oven-drying, steaming, shredding, and steeping, and combina-
tions of these methods, to reduce the cyanogenic glycoside content. Table 5 presents
HCN and phenolic content expressed as phytate acid and tannic acid. The combina-
tion of shredding and sun-drying was the most effective technique to reduce the
HCN content.
Table 4 Distribution of the phenolics and total potential cyanide by the parts of the cassava root
used for food
Dehydrated cassava leaves
Compounds found Dry in oven Dry by lyophilization
Phenolics (g 100 g−1) 0.65 2.66
Tannins (g 100 g−1) 0.10 0.90
Phytic acid (g 100 g−1) 0.35 0.51
Potencial cyanide (mg kg−1) 85.17 269.39
Source: Ortega-Flores et al. [70]
636 M. P. Cereda
Table 5 Distribution of hydrocyanic acid (HCN) and polyphenols in cassava leaves, expressed as
Phytate and Tannic acid
HCN Phytate Tannic acid
Variedades mg 100 g−1 g 100 g−1
MS 6 56.5 249.1 7.5
TSM 30555 40.2 197.8 15.0
TSM 30572 54.1 213.8 9.2
Ege Oda 60.6 107.3 6.9
Source: Fasuyi [71]
The effects of phenolic compounds are many, but for the scope of this chapter the
anticancer effect is highlighted, as a complement to the effects of cyanogenic
compounds.
Kyselova [62] points out that as a control mechanism of cancer cells, the proper-
ties of some phenolic compounds to induce the apoptosis of tumor cells and antican-
cer activity of pro-oxidant phenoxyl radicals stand out. Apoptosis is the mechanism
of programmed cell death, necessary to maintain a balance between cell prolifera-
tion and cell loss. Poor regulation of this balance can lead to malignant transforma-
tion, while induction of apoptosis suppresses cancer development [73, 74]. Several
diet-derived compounds, e.g., resveratrol, have been shown to induce apoptosis in
malignant cells and provide a promising natural strategy for preventing cancer [67,
75–80]. In general, the anticancer activity is linked to the antioxidant activity of the
phenolic compounds, which characterize all the phenolic compounds identified in
the cassava plant [67].
Specifically with respect to the resveratrol phenolic compounds present in red
grape skins, Bhat and Pezzuto [73] point out that cancer chemo preventive agents
reduce the incidence of tumorigenesis by intervening at one or more stages of car-
cinogenesis. Resveratrol was first shown to act as an antioxidant and antimutagenic
agent, thus acting as an anti-initiation agent, but it has also been established that it
acts to suppress and inhibit the formation of carcinogen-induced preneoplastic
lesions.
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 637
Of the phenolic compounds identified in the cassava plant, only scopoletin, from
the Coumarin group, was specifically mentioned among the phenolic compounds of
cassava. Scopoletin is a phenolic compound identified in the cassava plant that may
have characteristics attributed to linamarin. Obidoa and Obasi [77] describe scopol-
amine as isolated and identified in gari, a fermented flour consumed in Nigeria. Its
content in gari and unfermented flours is not altered by sun drying, refrigeration,
and storage processing, unlike what happens with linamarin. Scopolamine has also
been identified as an active ingredient in fruit infusions of Tetrapleura tetraptera,
used as medicine in East Africa. It is a potent hypotensive and nonspecific spas-
modic agent. These pharmacological effects of scopolamine are likely underlying
factors in the effect showed in tropical neuropathy characterized by optic nerve
atrophy, dizziness of nervous origin, and ataxia endemic among the population,
which consumes cassava and gari as subsistence foods. These symptoms are usually
attributed to cyanogenic glycosides present in cassava.
Ezeanyika et al. [78] describe the effects of scopoletin (6-methoxy-7-hydroxy-
coumarin) compared to cyanide on female Wistar rats serum Na+, K+, urea, creati-
nine, and some hematological parameters. Rats were randomly given doses of
7 g ml−1, 21 g ml−1, 35 g ml−1 scopoletin, 1.8 mg ml−1 cyanide, 10% dimethyl sulf-
oxide (the vehicle for the administration of scopoletin), and 1 ml distilled water
orally per kilogram of body weight at 24-h intervals. Scopoletin at the level found
in processed cassava diet (7 g ml−1) increased symptoms equivalent to 1.8 mg ml−1
cyanide, which is approximately the level consumed on a daily basis by a 70-kg man
in cassava-consuming populations. Among the groups administered with scopole-
tin, the serum levels of K+ and creatinine increased with increasing concentration of
scopoletin, while Na+ and Urea level were decreased. The results of this study sug-
gest further investigations on the effects of scopoletin and cyanide on hematopoiesis
and kidney function.
In addition to scopoletin, other phenolics present in the cassava plant have effects
very similar to the effects attributed to cyanide. Curcumin, a dietary phenolic com-
pound, also showed such effect by opening high conductance permeability transi-
tion pores in mitochondria, through initiates onset and induced mitochondrial
swelling, calcium release, impaired respiration, and through decreased mitochon-
drial membrane potential [79].
Tannins are also present in cassava. Rickard [63] reports the presence of low
levels of tannins (proanthocyanidins) detected in dried cassava samples using stan-
dard methods of analysis based on extraction procedures. The direct vanillin assay
method was compared with other direct methods of analysis based on acid hydroly-
sis and protein precipitation. The specificity of the methods is discussed in relation
to the levels of tannins detected and their potential biological activity in freeze-dried
and dried/processed samples. The results are discussed with respect to the amount
of indigestible material present in the samples. The results of these experiments
indicate that tannins present in dried/processed cassava products may be a factor
limiting their nutritional value. This characteristic, duly controlled, is nowadays
considered a positive characteristic, in view of the need to control excess weight,
which is increasing among adults and children.
638 M. P. Cereda
General data from in vitro and in vivo laboratory studies, epidemiological inves-
tigations, and human clinical trials indicate that phenolic compounds are beneficial
to human health. A large number of studies have identified cellular targets of plant
phenolic compounds in the diet that could be involved in health-promoting actions.
They also point out that dietary supplements are not classified as drugs and do not
require Food and Drug Administration (FDA) approval and that this is a sufficient
reason to evaluate and consider potential toxicities and drug interactions of phenolic
compounds [61].
3 Dietary Fiber
Awareness of the need to promote health and well-being has led to growing demand
for healthy food consumption, which research has shown to be intrinsically linked
to dietary habits [79]. The positive effects of dietary fiber are partly related to the
fact that a portion of its components go through a fermentation process that takes
place in the large intestine, which impacts the speed of intestinal transit, the pH of
the colon, and the production of byproducts with important physiological functions.
Individuals with high fiber intake appear to be at lower risk for developing coronary
heart disease, hypertension, obesity, diabetes, and colon cancer. Increased fiber
intake reduces serum cholesterol levels, improves blood glucose in patients with
diabetes, reduces body weight, and has been associated with lower serum levels of
ultrasensitive C-reactive protein. Fiber consumption of more than 14 g 1000 kcal−1
is generally regarded to confer health benefits, including the reduction of low-grade
inflammatory processes. The consumption of more fiber triggers numerous benefi-
cial effects, reducing by up to 27% the risk of nondegenerative chronic diseases
such as type 2 diabetes mellitus and other pathologies such as cardiovascular dis-
ease and colon cancer [81]. Therefore, it can be said that, although dietary fibers do
not have a curative effect, they can prevent or improve conditions for coping with
diseases and are therefore treated as another health-promoting factor [6]. According
to Ortega-Flores et al. [70], fibers are part of the cell walls of plants, and in humans,
they are responsible for regulating the intestinal tract and weight, carbohydrate and
lipid metabolism, and colon functioning.
The desirable properties of fibers and resistant starches differ with botanical
sources. These include swelling, gelation, increased viscosity, and water-binding
capacity [7–81].
Classified as soluble or not in water, insoluble fiber can help control constipation,
diverticular disease, atherosclerosis, and cardiovascular diseases. Soluble fiber con-
trols cholesterol and insulin metabolism. However, excess fiber in the diet can
reduce the absorption of nutrients in the intestine due to the speed of transit [6].
The solubility and insolubility of dietary fibers determine their physiological
effects when ingested in the diet. Soluble fibers have the ability to increase stomach
viscosity, reducing postprandial blood glucose and cholesterol levels. Therefore,
they interfere with the ability to retain water, accelerate intestinal transit, increase
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 639
fecal volume, and stimulate colonic fermentation, in addition to reducing the risk of
diseases of the gastrointestinal tract [82].
Understanding of the physiological importance of dietary fiber has increased
considerably over the last 10 years. The degree of indigestibility of the fibers con-
tained in foods, of which they are an integral part, can prevent some gastrointestinal
and metabolic diseases. They pass through the digestive tract without modification
and increase the stool, stimulating and making bowel movements more frequent. In
general, they remain in the gut environment longer, with important interference in
peristalsis performance [83].
In addition to this more mechanical importance, more recently, functional prop-
erties have been added to their positive effects, which include the ability to retain
water by increasing fecal volume, increasing intestinal viscosity, and controlling
intestinal transit time by increasing or decreasing it, with the consequent influence
on nutrient absorption [81].
In addition to other benefits, fiber promotes the reduction of digestion and
absorption of macronutrients, reduces the levels of serum lipids and postprandial
glucose, promoting glycemic control.
The effects of fiber are mainly attributed to the degree of viscosity and fermenta-
tion [81–85]. They produce short-chain fatty acids (SCFAs), mainly propionic acid,
when they are fermented in the large intestine by intestinal bacteria. In in vitro tests,
this generation of short-chain fatty acids, such as butyrate, has been associated with
benefits for gut health, with the modulation of cell proliferation, apoptosis, and the
regulation of angiogenesis [86]. The type of fatty acid formed varies with the dietary
fiber consumed [85], which results in the importance of evaluating the dietary fibers
available on the market and those potentially available for consumption.
In addition to being available, dietary fibers have evaluated for their performance.
Fiber characterization is vital for food manufacturers so that they can provide valid
and accurate information on product labeling. It is also fundamental for the activity
of the regulatory authorities regarding claims of nutritional and health properties.
Such information is still necessary for consumers, who use nutritional information
declared on food labels and associated materials.
Among the components of dietary fiber and its associated compounds, the phe-
nolic compounds that were included as a result of changes in the concept of healthy
foods stand out. These are secondary compounds with important beneficial func-
tions for the human body, preventing injuries caused by free radicals, which are
oxidized by them and are more stable. The biological effects of these molecules are
related to anti-inflammatory, anticarcinogenic activity and reduced incidence of car-
diovascular diseases [87, 88].
The fact that it has a high content of crude fiber does not mean that the raw mate-
rial is a source of fiber of good nutritional quality. Fiber is considered a functional
food and performs important functions in the body, such as promoting a feeling of
satiety, facilitating with the metabolism of lipids and carbohydrates and the physiol-
ogy of the gastrointestinal tract, and ensuring a slower absorption of nutrients [89].
For Bernaud and Rodrigues [90] the positive effects of dietary fiber are related,
in part, to the fact that a portion of the fermentation of its components occurs in the
640 M. P. Cereda
large intestine, which has an impact on the speed of intestinal transit, on the pH of
the colon, and on the production of byproducts with an important physiological
function. Individuals with high fiber intake appear to be at lower risk for developing
coronary heart disease, hypertension, obesity, diabetes, and colon cancer. Increased
fiber intake reduces serum cholesterol levels, improves blood glucose in patients
with diabetes, reduces body weight, and has been associated with lower serum lev-
els of ultrasensitive C-reactive protein. Higher fiber intake and intake of more fiber
in proportion to the energy consumed, currently recommended at 14 g 1000 kcal−1,
may bring greater health benefits, including the reduction of low-grade inflamma-
tory processes. The authors conclude that to meet increased need for fiber, the intake
of at least 3 g day−1 of fiber is necessary for the benefits to be realized, requiring the
consumption of a wide variety of sources of fiber, such as fruits, vegetables, whole
grains, and bran.
However, in modern life, these recommendations are not always easily met,
especially regarding the amount of fiber to be consumed daily. For this reason, spe-
cialized stores and gyms value, make available, and feature these products as food
supplements and encourage the use of new ranges of products such as oat bran, rice
bran, corn, wheat and passion fruit fiber, coconut flour, and eggplant, among others.
In response to this demand, food companies have been developing functional
food formulations, capable of meeting nutritional needs and delivering health ben-
efits, without losing sight of the needs of modern life; however, for these foods to be
considered health-promoting, there is a need to prove their beneficial
characteristics.
Among all sources of dietary fiber, cereal bran and rice and wheat bran are the
most studied and popular dietary supplements. Specific studies on the phytochemi-
cal composition of rice bran have proven its richness in phenolic compounds, vita-
mins with an emphasis on vitamin E, dietary fiber, oils, steroid derivatives,
polysaccharides, antioxidants, and proteins (essential amino acids, especially
lysine). For wheat bran, in addition to fiber, lignin and lignans, oligosaccharides,
polyphenols, phytic acid, minerals, alkyl resorcinols, glutathione, sulfur com-
pounds, α-linoleic acid, carotenoids, and vitamins B and E [91]. Wheat bran is
known and consumed globally. It is an important byproduct of cereal production,
especially when the grain has significant amounts of fermentable carbohydrates and
dietary fiber, in addition to resistant starch. About 50% fiber can be found in its
composition, 40% of which is hemicellulose, which represents a strong indication
of being a good source of dietary fiber [92, 93], considered a standard fiber [94].
Wheat fiber is easily found in Brazil and stands out globally as a relevant byproduct
of cereal production [95]. Due to its valorization, wheat fiber fetches high prices, so
other residues rich in fibers produced from Brazilian raw materials have been stud-
ied with a view to its valorization, including cassava bran.
Cassava fibers are considered compatible with the concept of dietary fiber.
According to the Brazilian Food Composition Table [3], raw cassava roots have up
to 2 g fiber/100 g, with 133 kcal, but with cooking it absorbs more water, and this
content reduces to 1.78, as does the caloric content (121 kcal) in the same 100 g. In
contrast, when dehydrated in the form of flour, the energy content rises to 160 kcal
and the dietary fiber content exceeds 5.0 g/100 g.
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 641
The bran obtained in the commercial extraction of cassava starch has been exten-
sively studied because it is abundant, obtained in good sanitary conditions, and
available at low cost. Its production on an industrial scale was quantified by Cereda
[1], at an average of 140 kg of dry mass per ton of processed cassava.
Based on a description by Leonel et al. [96] in the characterization of cassava
bran from the point of view of dietary fiber, the authors highlighted that 80% of
residual starch would provide a very high calorie content when consumed, as glu-
cose predominated in this sample, followed by of 22.3% hemicellulose. Discounting
the starch, we arrive at 58% fiber, 4% protein, and 6% ash.
By the year 2000, this content had already been reduced to 30–50% starch on a
dry weight basis. The authors comment that this fibrous material, a byproduct of
cassava processing, may be valuable thanks to its rich organic nature and low ash
content, as an ideal substrate for microbial processes for the production of value-
added products. Attempts have been made to make several products from cassava
bran, such as organic acids, flavor and aroma compounds, and mushrooms [97].
Silva et al. [98] characterized cassava bran and reported a caloric value of 179.32 kcal
due to the presence of residual starch, reduced by the use of amylolytic and cellulo-
lytic enzymes, with a consequent increase in its functionality and its value as a
source of dietary fiber. Later, Lacerda [99] compared cassava bagasse fiber with
wheat fiber, a commercial and standard product for dietary fiber (Tables 6 and 7).
In general appearance, commercial wheat bran and cassava bran are similar, but
wheat fiber is darker, whereas cassava bran is more red and yellow. As sources of
dietary fiber, commercial fiber and cassava fiber are very comparable. Wheat bran
has a much higher phenolic content. In terms of quality, a higher rate of water and
oil absorption is observed for cassava fiber and, surprisingly, greater water solubil-
ity for wheat fiber. The results presented only highlight the potential of finding qual-
ity fiber in tropical raw materials.
Table 6 Aspect and color of wheat fiber (Yoki®), arrowroot and cassava bran and standard
deviation of the mean
Wheat fiber Cassava bran
General aspect
Brightness (L%) 52.65 58.51
Red 7.26 4.40
Yellow 18.30 16.53
Source: Lacerda et al. [99]
Averages of three repetitions
642 M. P. Cereda
Table 7 Physicochemical characterization and nutritional aspects of wheat fiber, arrowroot and
cassava bran
Análises Unities [DM] Wheat fiber Cassava bran
Sugars
-TRS g 100 g−1 26.37 29.16
-RS g 100 g−1 3.54 7.42
Protein g 100 g−1 6.90 1.80
Fat g 100 g−1 1.8 2.37
Calorific value Kcal 100 g−1 149c 144b
Ash g 100 g−1 2.62 1.44
FDN g 100 g−1 60.04 60.04
FAD g 100 g−1 12.91 29.22
Phenolics μg 100 g−1 29.04 1.53
Dietary fiber total g 100 g−1 46.21 44.32
Functional properties
WAI 3.85 4.92
OAI 2.84 3.36
WSI 12.20 1.73
Source: Lacerda et al. [99]
Averages of three replicates expressed in dry mass
TRS Total Reducing Sugars, RS Reducing Sugars, FND Fiber by acid detergent, FAD Fiber by
neutral detergent, FAT Dietary fiber total, WAI water absorption index, OAI oil absorption index,
WSI water solubility index
4 Final Considerations
the possibility that they will be generated. In the specific case of scopoletin, it
would be important to verify to what extent it can be an origin or an adjuvant in
chronic diseases such as Konzo, which is generally attributed to linamarin.
• On the potential of cassava root fiber as a phytotherapy
• The evaluation of cassava products (e.g., roots, leaves, starch, industrial resi-
dues) as dietary fibers is even less researched than cyanogenic compounds and
phenolic compounds. The available information shows potential for cassava
bagasse, as it is a safe and abundant industrial waste.
• To guide the complementary research needed to configure the cyanogenic com-
pounds present in the cassava plant as a phytotherapeutic agent with an effect on
cancer, it would be important to relate cassava consumption per inhabitant/year
with diagnosed cases of cancer, especially with cancer of the digestive system,
with an emphasis on the intestine, which is where cyanide would be released and
absorbed.
References
1. Cereda MP (2002) Characterization of starchy raw materials. In: Cereda MP, Vilpoux OF
(Org). General properties of starch, 1st edn. Fundação Cargill: São Paulo, Livro 1, v.1,
pp 88–133. (In Portuguese)
2. Sagrilo E, Pequeno MG, Vidigal Filho P, Scapim C (2002) Effect of harvest time on vegeta-
tive growth, yield and root quality of three cassava cultivars. Bragantia 61(August):115–125.
https://doi.org/10.1590/S0006-87052002000200005. (In Portuguese)
3. TACO- Brazilian Food Composition Table (2011) Núcleo de Estudos e Pesquisas em
Alimentação, Universidade Estadual de Campinas, 4 edn. NEPA-UNICAMP: Campinas, 161
p. (In Portuguese). Avaiable in: http://www.fcf.usp.br/tbca/. Consulted in January 3th/2021.
(In Portuguese)
4. Cressey P, Reeve J (2019) Metabolism of cyanogenic glycosides: a review. Food Chem
Toxicol 125:225–232. https://doi.org/10.1016/j.fct.2019.01.002. PMID: 30615957
5. Gensa U (2019) Review on cyanide poisoning in ruminants. J Biol Agric Healthcare
9(6):1–12. https://doi.org/10.7176/JBAH
6. Fuller S, Beck E, Salman H et al (2013) New horizons for the study of dietary fiber
and health: a review. Plant Foods Hum Nutr 43(1):408–416. https://doi.org/10.1590/
S1983-40632013000400005
7. Fuentes-Zaragoza E, Riquelme-Navarrete MJ, Sánchez-Zapata E, Pérez-Álvarez JA (2010)
Resistant starch as functional ingredient: a review. Food Res Int 43(4):931–942. https://doi.
org/10.1016/j.foodres.2010.02.004
8. Sykes AH (1981) Early studies on the toxicology of cyanide. Cyanide Biol:1–9
9. Seigler DS (1991) Cyanide and cyanogenic glycosides. In: Rosenthal GA, Berenbaum MR
(eds) Herbivores: their interactions with secondary plant metabolites. Academic Press, San
Diego, pp 35–77
10. Conn EE (1978) Cyanogenesis, the production of hydrogen cyanide, by plants. In: Effects of
poisonous plants on livestock. Academic Press, pp 301–310. https://doi.org/10.1016/B978-0
-12-403250-7.50035-2
11. Hösel W (1981) Cyanide in biology. Academic Press, London, pp 217–232
644 M. P. Cereda
12. Møller BL, Seigler DS (1999) Biosynthesis of cyanogenic glycosides, cyanolipids and related
compounds. In: Plant amino acids biochemistry and biotechnology. CRC Press, pp 563–609
13. FAO/WHO (1991) Joint FAO/WHO food standards programme. Codex Alimentarius
Commission XII, Suppl. 4, FAO, Rome
14. EFSA (2004) Evaluation of the health risks related to the presence of cyanogenic glycosides in
foods others than raw apricot kernels. J EFSA:89. https://doi.org/10.2903/j.efsa.20YY.NNNN
15. Nambisan B (2011) Strategies for elimination of cyanogens from cassava for reducing toxic-
ity and improving food safety. Food Chem Toxicol 49(3):690–693. https://doi.org/10.1016/j.
fct.2010.10.035
16. Onyenwoke CA, Simonyan KJ (2014) Cassava post-harvest processing and storage in Nigeria:
a review. Afr J Agric Res 9(53):3853–3863. https://doi.org/10.5897/AJAR2013.8261
17. Knudsen I, Søborg I, Eriksen F, Pilegaard K, Pedersen J (2008) Risk management and
risk assessment of novel plant foods: concepts and principles. Food Chem Toxicol
46(5):1681–1705. https://doi.org/10.1016/j.fct.2008.01.022
18. Rosling H (1988) Cassava toxicity and food security. A review of health effects of cyanide
exposure from cassava and of ways to prevent these effects, 40 p. ISBN: 91-971029-0-3
19. Union of Pure and Applied Chemists. Available in https://iupac.org/who-we-are/. Consulted
in 24th February 2022
20. Bokanga M, Essers S, Pouler N, Rosling H, Tewe O, Asiedu R, Brader L (1994) Preface. Acta
Horticult, Wageningen 375:1–2
21. Day AJ, DuPont MS, Ridley S, Rhodes M, Rhodes MJ, Morgan MR, Williamson G
(1998) Deglycosylation of flavonoid and isoflavonoid glycosides by human small intestine
and liver β-glucosidase activity. FEBS Lett 436(1):71–75. https://doi.org/10.1016/
S0014-5793(98)01101-6
22. Nartey F (1981) Cyanogenesis in tropical feeds and foodstuffs. In: Vennesland B, Conn
EE, Knowles CJ, Westley J, Wissing F (eds) Cyanide in biology. Academic Press, London,
pp 115–132
23. De Bruijn GH (1973) Cyanogenic character of cassava (Manihot esculenta). In: Chronic cas-
sava toxicity. IDRC, Ottawa
24. Bokanga M (1992) Contraints in food and nutrition research. In: Thottapplly G, Monti LM,
Mohan Raj DR, Moore AW (eds) Biotechnology: enhancing research on tropical crops in
Africa. International Institute of Tropical Agriculture, Ibadan, pp 33–38
25. Oluwole OSA, Onabolu AO, Mtunda K, Mlingi N (2007) Characterization of cassava (Manihot
esculenta Crantz) varieties in Nigeria and Tanzania, and farmers’ perception of toxicity of
cassava. J Food Compos Anal 20(7):559–567. https://doi.org/10.1016/j.jfca.2007.04.004
26. Cardoso Júnior NDS, Viana AES, Matsumoto SN, Sediyama T, Carvalho FMD (2005)
Effect of nitrogen on agronomic characteristics of cassava. Bragantia 64(4):651–659. (In
Portuguese)
27. Bourdoux P, Seghers P, Mafuta M, Vanderpas J, Vanderpas-Rivera M, Delange F et al (1982)
Nutritional factors involved in the goitrogenic action of cassava. IDRC, Ottawa
28. Boakye Peprah B, Parkes EY, Harrison OA, van Biljon A, Steiner-Asiedu M, Labuschagne
MT (2020) Proximate composition, cyanide content, and carotenoid retention after boiling of
provitamin A-rich cassava grown in Ghana. Foods 9(12):1800
29. EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin
L, Chipman JK, del Mazo J et al (2019) Evaluation of the health risks related to the presence
of cyanogenic glycosides in foods other than raw apricot kernels. EFSA J 17(4):e05662.
https://doi.org/10.2903/j.efsa.2019.5662
30. Zhu C, Krumm C, Facas GG, Neurock M, Dauenhauer PJ (2017) Energetics of cellulose
and cyclodextrin glycosidic bond cleavage. React Chem Eng 2(2):201–214. https://doi.
org/10.1039/C6RE00176A
31. Cagnon JR, Cereda MP, Pantarotto S (2002) Cassava cyanogenic glycosides: biosynthesis,
distribution, detoxification and dosage methods. In: Agricultura: tuberosas amiláceas Latino
Americanas, vol 2. Fundação Cargill, São Paulo, pp 83–99. (In Portuguese)
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 645
32. Ramalho RT, Aydos RD, Cereda MP (2010) Evaluation of acetone cyanohydrin effect in “in
vitro” inativation of the Ehrlich ascites tumor cells. Acta Cir Bras 25(1):111–116. https://doi.
org/10.1590/S0102-8650201400140008
33. Hughes J, de Carvalho JPC, Hughes MA (1994) Purification, characterization, and cloning
of α-hydroxynitrile lyase from cassava (Manihot esculenta Crantz). Arch Biochem Biophys
311(2):496–502. https://doi.org/10.1006/abbi.1994.1267
34. Idibie CA, Davids H, Iyuke SE (2007) Cytotoxicity of purified cassava linamarin to a
selected cancer cell lines. Bioprocess Biosyst Eng 30(4):261–269. https://doi.org/10.1007/
s00449-007-0122-3
35. Orjiekwe CL, Solola A, Iyen E, Imade S (2013) Determination of cyanogenic glucosides in
cassava products sold in Okada, Edo State, Nigeria. Afr J Food Sci 7(12):468–472. https://
doi.org/10.5897/AJFS2013.1012
36. FAO/WHO (2019) Discussion paper on the establishment of MLs for HCN in cassava and
cassava-based products and occurrence of mycotoxins in these products. Joint FAO/WHO
Food Standards Programme Codex Committee on Contaminants in Foods. 13th Session.
Yogyakarta, Indonesia, 29 April – 3 May 2019: [s.n.]. Disponível em: https://www.google.
com/search?client=firefox-b-d&sxsrf=ALeKk00yvKwTdmWTDiTHE04CytocXyRm_A:16
12304652672&q=datatable+FAO/WHO+(1988)+established+10+mg+HCN/kg+live+weigh
t&spell=1&sa=X&ved=2ahUKEwiJ_N3wnszuAhXJHLkGHbB1AqwQBSgAegQIBxA1&b
iw=1366&bih=666. Consultado em 2 Fev. 2021
37. Philbrick DJ, Hill DC, Alexander JC (1977) Physiological and biochemical changes associ-
ated with linamarin administration to rats. Toxicol Appl Pharmacol 42(3):539–551. https://
doi.org/10.1016/S0041-008X(77)80039-2
38. EFSA Journal (2007) Opinion of the scientific panel on contaminants in the food chain
on a request from the commission related to cyanogenic compounds as undesirable sub-
stances in animal feed. EFSA J 434:1–67. www.efsa.europa.eu Page 1 of 67 Question N°
EFSA-Q-2003-064 Adopted on 23 November 2006
39. Logsdon MJ, Hagelstein K, Mudder T (1999) The management of cyanide in gold extraction.
International Council on Metals and the Environment, Ottawa, p 10
40. Tomasik P, Jane JL, Spence K, Andernegg JW (1995) Starch ferrates. Starch-Stärke
47(2):68–72. https://doi.org/10.1002/star.19950470207
41. FAO/WHO (2013) Proposed draft maximum levels for hydrocyanic acid in cassava and cas-
sava products. Codex Committee on Contaminants in Foods. Joint FAO/WHO Food Standards
Programme Codex Committee on Contaminants in Foods. Seventh Session, Moscow,
Russian Federation, 8–12, April, 2013: [s.n.]. Disponível em: https://www.google.com/url?s
a=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwiR-sXynszuAhUpD7kGHRSPD
b8QFjAAegQIAxAC&url=https%3A%2F%2F2.zoppoz.workers.dev%3A443%2Fhttp%2Fwww.fao.org%2Ftempref%2Fcodex%2FMeet
ings%2FCCCF%2Fcccf7%2Fcf07_10e.pdf&usg=AOvVaw1e6FYAdKN0UmRow_sEKa6i.
Consultado em 2 Fev. 2021
42. Poulton JE (1993) Cyanogenic compounds in plants and their toxic effect. In: Keler RF, Tu
AT (eds) Handbook of natural toxins, Plant and fungal toxins, vol v. 1. M. Dckker, New York,
pp 117–157
43. Nelson L (2006) Acute cyanide toxicity: mechanisms and manifestations. J Emerg Nurs
32(4):S8–S11. https://doi.org/10.1016/j.jen.2006.05.012
44. Cheeke PR (1989) Toxicants of plant origin: alkaloids (v.1). CRC Press, Boca Raton. ISBN
0-8493-6990-8
45. Baskin SI, Kurche JS, Maliner BI (2004) Cyanide. In: Roy MJ (ed) Physician’s guide to ter-
rorist attack. Humana Press, Totowa. https://doi.org/10.1007/978-1-59259-663-8_19
46. Ballantyne B (1987) Toxicology of cyanides. In: Ballantyne B, Marrs TC (eds) Clinical and
experimental toxicology of cyanides. IOP Publishing Limited, Bristol, pp 41–126
47. Burtis CA, Ashwood ER (1996) Enzymes. In: Teitz fundamentals of clinical chemistry. NB
Saunders Company, Philadelphia, pp 4312–4335
646 M. P. Cereda
48. Essers AJA (1994) Further improving the enzymic assay for cyanogens in cassava products.
Acta Horticult 375:97–101
49. Oke OL (1969) The role of hydrocyanic acid in nutrition. World Rev Nutr Diet 11:170–198.
https://doi.org/10.1159/000387578
50. Diniz M, de Mattos MCY, Cereda MP (1996) Linamarin: the toxic compounds of cassava. J
Venomous Toxins 2(1):06–12
51. Piste P (2013) Cysteine–master antioxidant. Int J Pharm Chem Biol Sci 3(1):143–149
52. Kashala-Abotnes E, Okitundu D, Mumba D, Boivin MJ, Tylleskär T, Tshala-Katumbay D
(2019) Konzo: a distinct neurological disease associated with food (cassava) cyanogenic poi-
soning. Brain Res Bull 145:87–91. https://doi.org/10.1016/j.brainresbull.2018.07.001
53. WHO World Health Organization (1993) Hydrogen cyanide and cyanides: human health
aspects. United Nations Environmental Programme, Geneve, 45 p
54. Burns AE, Bradbury JH, Cavagnaro TR, Gleadow RM (2012) Total cyanide content of cas-
sava food products in Australia. J Food Compos Anal 25:79–82. https://doi.org/10.1016/j.
jfca.2011.06.005
55. Chisté RC, Cohen KO (2008) Determination of total cyanide in cassava flour of the
dry and water group commercialized in the city of Belém-PA. Revista Brasileira
de Tecnologia Agroindustrial 2(2):96–102. Avaiable in: https://doi.org/10.3895/
S1981-36862008000200010. Consulted in: February 16th 2022. (In Portuguese)
56. Oliveira OSD, Brito VHDS, Cereda MP (2018) Establishing a standard for handmade
Brazilian cassava flour from Baixada Cuiabana (Mato Grosso, Brazil) to support its process-
ing and sale. Food Sci Technol 39:559–566. https://doi.org/10.1590/fst.30117
57. Sulyok M, Beed F, Boni S, Abass A, Mukunzi A, Krska R (2015) Quantitation of multiple
mycotoxins and cyanogenic glucosides in cassava samples from Tanzania and Rwanda by an
LC-MS/MS-based multi-toxin method. Food Addit Contam Part A 32(4):488–502. https://
doi.org/10.1080/19440049.2014.975752
58. Joint FAO/WHO (2016) Expert Committee on Food Additives. Meeting, & World Health
Organization. Evaluation of Certain Food Additives and Contaminants: Eightieth Report of
the Joint FAO/WHO Expert Committee on Food Additives, vol 80. World Health Organization
59. Expert Committee on Food Additives (JECFA) (2016) World Organization for Health.
Disponivel em: https://www.who.int/foodsafety/chem/jecfa/ARfd/en/. Consultado em 21
Fevereiro 2021
60. Food and Agriculture Organization of the United Nations, Codex Alimentarius Commission
(2007) Cereals, pulses, legumes and vegetable proteins, 1st edn. Committee Food and
Agriculture Organization of the United Nations, Rome. Retrieved from http://www.fao.
org/3/a-a1392e.pdf
61. Shahidi F, Naczk M (1995) Food phenolics: sources, chemistry, effects and aplications. Dent
Tech 37(1/2):75–107
62. Kyselova Z (2011) Toxicological aspects of the use of phenolic compounds in disease preven-
tion. Interdiscip Toxicol 4(4):173–183. https://doi.org/10.2478/v10102-011-0027-5
63. Rickard JE (1986) Tannin levels in cassava, a comparison of methods of analysis. J Sci Food
Agric 37(1):37–42. https://doi.org/10.1002/jsfa.2740370106
64. Padmaja G (1989) Evaluation of techniques to reduce assayable tannin and cyanide in cas-
sava leaves. J Agric Food Chem 37(3):712–716
65. Ferraro V, Piccirillo C, Tomlins K, Pintado ME (2016) Cassava (Manihot esculenta Crantz)
and yam (Dioscorea spp.) crops and their derived foodstuffs: safety, security and nutritional
value. Crit Rev Food Sci Nutr 56(16):2714–2727
66. Uarrota VG, Moresco R, Schmidt EC, Bouzon ZL, da Costa Nunes E, de Oliveira Neubert
E et al (2016) The role of ascorbate peroxidase, guaiacol peroxidase, and polysaccharides
in cassava (Manihot esculenta Crantz) roots under postharvest physiological deterioration.
Food Chem 197:737–746. https://doi.org/10.1016/j.foodchem.2015.11.025
Phitotherapeutic Potential of Cassava (Manihot esculenta, Crantz) 647
85. Kaczmarczyk MM, Miller MJ, Freund GG (2012) The health benefits of dietary fiber: beyond
the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer.
Metabolism 61(8):1058–1066. https://doi.org/10.1016/j.metabol.2012.01.017
86. Phillips GO, Cui SW (2011) An introduction: evolution and finalisation of the regula-
tory definition of dietary fibre. Food Hydrocoll 25(2):139–143. https://doi.org/10.1016/j.
foodhyd.2010.04.011
87. Bruneton J (1999) Pharmacognosy & phytochemistry medicinal plants. In: Techniques &
documentation, vol 81, 2nd edn. Lavoisier Publishers, Paris, pp 106–109
88. Kuskoski EM, Asuero AG, Parilla MCG, Troncoso AM, Fett R (2004) Antioxidant activity in
anthocianics pigments. Ciencia e Tecnologia de Alimentos (Brazil)
89. Alvarez T, Magnoni D, Cukier C (2005) Skin nutrition and healing. In: Nutrition based on the
physiology of organs and systems, pp 8–15. (In Portuguese)
90. Bernaud FSR, Rodrigues TC (2013) Dietary fiber: adequate intake and metabolism health
effects. Arquivos Brasileiros de Endocrinologia & Metabologia 57:397–405. https://doi.
org/10.1590/S0004-27302013000600001. (In Portuguese)
91. Sharif MK, Butt MS, Anjum FM, Khan SH (2014) Rice bran: a novel functional ingredient.
Crit Rev Food Sci Nutr 54(6):807–816. https://doi.org/10.1080/10408398.2011.608586
92. Yuan X, Wang J, Yao H, Chen F (2005) Free radical-scavenging capacity and inhibitory activ-
ity on rat erythrocyte hemolysis of feruloyl oligosaccharides from wheat bran insoluble dietary
fiber. LWT-Food Sci Technol 38(8):877–883. https://doi.org/10.1016/j.lwt.2004.09.012
93. Wang J, Sun B, Cao Y, Wang C (2010) In vitro fermentation of xylooligosaccharides from
wheat bran insoluble dietary fiber by Bifidobacteria. Carbohydr Polym 82(2):419–423.
https://doi.org/10.1016/j.carbpol.2010.04.082
94. Raupp DDS, Rosa DA, Marques SHDP, Banzatto DA (2004) Digestive and functional prop-
erties of a partially hydrolyzed cassava solid waste with high insoluble fiber concentration.
Sci Agric 61(3):286–291
95. Giuntini EB, Lajolo FM, Menezes ED (2003) Dietary fiber potential in Ibero-American coun-
tries: food, products and residues. Arch Latinoam Nutr 53(1):14–20. (In Portuguese)
96. Leonel M, Cereda MP, Roaux X (1998) Cassava bagasse as a dietary food product. Trop
Sci 38(4):224–228. Available in: http://hdl.handle.net/11449/65385. Consulted in: 18th
February 2022
97. Pandey A, Soccol CR, Nigam P, Soccol VT, Vandenberghe LPS, Mohan R (2000)
Biotechnological potential of agro-industrial residues. II: cassava bagasse. Bioresour Technol
74(1):81–87. https://doi.org/10.1016/S0960-8524(99)00143-1
98. Silva ÉC, Cereda MP, Colman TD, Demiate IM, Schnitzler E (2015) Characterisation of
cassava bagasse in different granulometries from two starch processing plants. J Microbiol
Biotechnol Food Sci 05(02):99–102. https://doi.org/10.15414/jmbfs.2015.5.2.99-102
99. Lacerda AM, de Camargo L, Cereda MP (2021) Potential of arrowroot bran from starch
extraction as a fiber source. (In Portuguese). Article in writing based on the Dissertation
(Master in Biotechnology, Catholic University of Campo Grande, MS, Brazil, 2015. (In
Portuguese)
100. Jones DA (1998) Why are so many food plants cyanogenic? Phytochemistry 47(2):155–162.
https://doi.org/10.1016/S0031-9422(97)00425-1
101. Behera SS, Ray RC (2017) Microbial linamarase in cassava fermentation. In: Microbial
enzyme technology in food applications. CRC Press, pp 333–346
102. McMahon JM, White WL, Sayre RT (1995) Cyanogenesis in cassava (Manihot esculenta
Crantz). J Exp Bot 46(7):731–741
103. Gomez S, Stuefer JF (2006) Members only: induced systemic resistance to herbivory in a
clonal plant network. Oecologia 147(3):461–468
Phytochemistry and Pharmacological
Studies of Indian Cinnamomum Schaeff
1 Introduction
Plants have been used for therapeutic purposes since the ancient times and about
400,000 plant species were reported around the world [1]. But only a small fraction
of these plant species, i.e., about 35,000–70,000, has been screened for their medici-
nal use [2]. India has a vast geographical area with high potential medicinal plants
used in Ayurveda, Sidha, Unani, and traditional medicines. The WHO reported that
of the 21,000 medicinal plants used all around the world, 2500 are found in India
[3]. The primary sources of medicine for early drug discovery are plants that are
reported to have ethno-pharmacological uses. Plant-derived compounds have better
patient tolerance and acceptance. Plant-derived compounds also have a long history
of clinical use [4] Many currently prescribed drugs were originally isolated from
plants and/or are semisynthetic analogues of phytochemicals [5].
The genus Cinnamomum belongs to the Lauraceae family consisting of 250 spe-
cies of trees and shrubs distributed in Southeast Asia, Australia, China, and Africa.
Most of Cinnamomum species are aromatic with a lot of medicinal and economic
importance as sources of essential oils, spices, and therapeutic drugs. Cinnamomum
species are widely used in herbal therapy in treating bronchitis, colds, sinusitis, and
fungal infections [6]. Their barks and leaves were used in foods as flavoring agent
and seasoning [7]. Several species of this genus such as C. malabatrum, C. walai-
warense, and C. trivancoricum were used to treat stomach pain. Cinnamomum
riparium, C. sulphuratum, C. filipedicellatum and C. wightii were used for treating
headaches, wounds, fever, and menstrual problems [8].
In traditional medicine, the cinnamon bark infusion was used as a remedy for
arthritis, rheumatism, nasopharyngeal infections, and stomach pain, whereas its
leaves, barks, and roots are used to treat diarrhea and dysentery [9], rheumatism and
inflammation [10], and neuralgic headaches. Mustaffa et al. [11] reported that the
leaves of C. iners are used to relieve fever and digestive problems and are used as
carminative [12]. Cinnamomum sulphuratum is reported to have anti-inflammatory
[13], hepatoprotective, and antimicrobial properties and is used for treating wounds,
fever/pyrexia, headache, backache [14], cholera, dyspepsia [15], menstrual prob-
lems, and worm infestation [16].
Cinnamomum zeylanicum leaf oil is used to treat toothache and its dried leaves
are used to induce menstruation [17] and also it has been used as a sweating agent
and an analgesic [18]. A wide range of pharmacological effects has been reported in
C. cassia including antitumor, anti-inflammatory, analgesic, neuroprotective, anti-
bacterial, antiviral, cardiovascular protective, immunoregulatory, antidiabetic, anti-
obesity, cytoprotective, and anti-tyrosinase effects. Barks of Cinnamomum
camphora are used as antispasmodic, anodyne, sedative, anthelmintic, diaphoretic,
stimulative, and carminative agents [19]. Moreover, barks of Cinnamomum malaba-
trum are used as carminative agents and are reported to have antispasmodic, astrin-
gent, antiseptic, hemostatic, stomachic, and germicidal properties. It is reported that
oil from the barks of Cinnamomum malabatrum has the ability to cure diarrhea,
cough, and dysentery, and its roots and leaves are used to treat rheumatism. The
plant has been known to have several pharmacological effects such as analgesic and
anti-inflammatory [20], antioxidant [21], and anticancer effects [22]. Kurokawa
et al. [23] reported that C. verum possesses significant antiulcerogenic, antiallergic,
anesthetic, and antipyretic activities. Barks and leaves of C. tamala are used as
stimulant and carminative agents to treat gonorrhea, rheumatism, and diabetes [24].
Phytochemicals are biologically active, naturally occurring chemical compounds
found in plants, which provide health benefits for humans [25]. They are found in
different parts of plants such as roots, stems, flowers, fruits, leaves, or seeds [26].
Bioactive compounds include an extremely heterogeneous class of compounds such
as tocopherols, polyphenolic compounds, phytosterols, carotenoids, and organosul-
fur compounds [27]. The present review aims to compile the detailed information
on phytocompounds and pharmacological properties reported in different species of
Cinnamomum in India.
Coumarin
Eugenol Camphor
component, and also moderate levels stem bark oils were detected in C. citriodorum
and C. sinharajanse. An unidentified Cinnamomum accession of Gammaduwa also
reported the presence of cinnamaldehyde. Rao et al. [38] isolated 25 compounds
from essential oils from the petiole of C. verum and carried out a GC-MS analysis.
The major components were (E)-cinnamaldehyde, eugenol, (E)-cinnamyl acetate,
and linalool.
Simic et al. [39] reported that the GC-MS analysis of C. verum detected eugenol,
cinnamaldehyde, cinnamaldehyde propylene, and limonene and a variety of terpe-
noid compounds (α-pinene, camphene). Mollenbeck et al. [40] reported a study on
C. verum essential oil. Trans-cinnamyl acetate was much higher in the flowers and
fruit volatile oils than in buds. The minor compounds included α-humulene and
α-muurolene. Leaf and bark oils of C. verum were rich in cinnamaldehyde [41] and
eugenol [42, 43]. Nath et al. [44] reported a chemotype of C. verum yielding benzyl
benzoate-rich leaf and bark essential oils from northeast India. The root-bark essen-
tial oil was reported to contain camphor as its main component in contrast to the
stem bark essential oil [45]. Linalool and (E)-cinnamyl acetate were the main con-
stituents of tender twigs’ essential oil [47].
Linalool, β-caryophyllene, and (E)-cinnamyl acetate were reported in essential
oils obtained from pedicels of buds, flowers, and fruits of C. verum [46, 47].
Mariappan et al. [48] analyzed chemical constituents of C. verum methanolic bark
extracted by GC-MS analysis. Trans-cinnamaldehyde, (E)-3-(2-methoxyphenyl)-2-
propenoic acid, 4-vinyl benzoic acid, and coumarin were the major chemical con-
stituents identified. Cinnamomum verum dried leaves collected from Delhi were
reported to contain 1,2-trans-sabinene hydrate, (Z)-β-ocimene, and germacrene A as
the major compounds and α-gurjunene, myrcene, α-pinene, and β-sabinene as the
minor compounds. Trans-sabinene hydrate, (Z)-β-ocimene, and germacrene A were
the chemotypes reported [49].
Kapoor et al. [50] reported eugenol as a significant constituent of C. verum dried
leaves collected from Gorakhpur, Uttar Pradesh. The minor constituents were
spathulenol, aromadendrene, viridiflorene, and methyl eugenol. Joshi et al. [51]
reported GC-MS analysis of fresh leaf oil collected from Jeolikote, Uttarakhand.
The oil contains (E)-cinnamaldehyde and linalool as major compounds and
(E)-cinnamyl acetate, β-pinene, and α-copaene as minor compounds. Chanotiya
et al. [52] reported the chemical constituents of C. verum from Nainital district,
Uttarakhand. (E)-Cinnamyl acetate, linalool, and (Z)-cinnamaldehyde were the sig-
nificant compounds isolated, whereas camphene, α-Pinene, 3-phenylpropanal,
benzaldehyde, bornyl acetate, (Z)-cinnamyl acetate, coumarin, salicylaldehyde, and
β-copaen-4α-ol were reported with meager amount.
Agrawal et al. [53] collected fresh aerial parts of C. verum samples from three
areas of Uttarakhand and analyzed their chemical compositions. Linalool and
(E)-cinnamaldehyde were the major constituents, and 1,8-cineole was the minor
constituent of samples collected from Munsiyari. Linalool, (E)-cinnamaldehyde,
and camphor were the major compounds of Lohaghat and Champawat samples.
Pithoragarh and Tanakpur samples were reported to contain significant compounds
such as linalool, (E)-cinnamaldehyde, and cinnamyl acetate. Eugenol,
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 655
The GC-MS studies of C. zeylanicum essential oil clearly showed the presence
of 38 components which include monoterpenes, sesquiterpenes, aromatic alde-
hydes, and ketones. Cinnamaldehyde was the major compound, followed by benz-
aldehyde [68]. Cinnamomum zeylanicum bark essential oil possesses compounds
such as cinnamic acid, cinnamaldehyde, eugenol, benzoic acid, benzaldehyde, trit-
erpenes, monoterpenes, and sesquiterpenes [69]. Vangalapati et al. [70] reported
presence of chemical constituents in different parts of C. zeylanicum. The barks and
leaves contain cinnamaldehyde and eugenol, respectively. Roots and barks showed
the presence of camphor and trans-cinnamyl acetate and the fruits β-caryophyllene.
Buds showed the presence of terpene hydrocarbons, alpha-bergamotene, alpha-
copaene, and oxygenated terpenoids. Flowers showed the presence of (E)-cinnamyl
acetate, trans-alphabergamotene, and caryophyllene oxide.
Jakhetia et al. [71] reported that C. zeylanicum contains cinnamic acid, cinnam-
aldehyde, cinnamate, trans-cinnamaldehyde, caryophyllene oxide, l-borneol,
l-bornyl acetate, eugenol, b-caryophyllene, E-nerolidol, cinnamyl acetate, terpin-
olene, a-terpineol, a-cubebene, and alpha-thujene. Cinnamomum zeylanicum oil has
been reported to contain chemical constituents such as cinnamic acid, benzoic acid,
and benzaldehyde whose lipophilic part is responsible for its antimicrobial proper-
ties [72]. Cinnamomum zeylanicum bark essential oil contains cinnamyl acetate
[73]. Brari and Thakur [74] reported cinnamaldehyde and linalool from the essential
oil isolated from C. zeylanicum. The essential oil of C. zeylanicum bark was rich in
trans-cinnamaldehyde [75].
Cinnamomum zeylanicum bud volatile oil has been reported to contain δ-cadinene,
tetradecanol, α-humulene, α-copaene, α-bergamotene, and viridiflorene. Leaf oil
contains (E)-cinnamaldehyde, eugenol, β-caryophyllene, linalool, (E)-cinnamyl
acetate, and α-terpineol. Moreover, fruit stalks oil contain α-humulene, caryophyl-
lene, (E)-cinnamyl acetate, δ-cadinene, α-copaene, and (E)-τ-cadinol. Flower oil of
C. zeylanicum contain trans-α-bergamotene, caryophyllene oxide, tetradecanal,
α-cadinol, and globulol. Similar enantiomeric distributions have been reported for
C. camphora essential oil [76]. Mallavarapu et al. [77] isolated essential oil of
C. zeylanicum collected from Bangalore and Hyderabad and analyzed it by using
GC and GC-MS. Eugenol was reported as the main constituent along with 47 other
constituents. Both oil samples were different with respect to the quantities of lin-
alool, (3-caryophyllene, (E)-cinnamaldehyde, (E)-cinnamyl acetate, and benzyl
benzoate. The main phytocompounds of oil collected from Bangalore were
a-phellandrene, eugenol, linalool, (E)-cinnamyl acetate (E)-cinnamaldehyde, and
P-caryophyllene, while those of oil collected from Hyderabad contained eugenyl
acetate, eugenol, benzyl benzoate, and linalool.
Mallavarapu and Ramesh [77] reported 49 constituents from fruit oil of C. zeyl-
anicum from Bangalore. The main constituents were a-pinene, P-caryophyllene,
G-cadinene, and a-muurolol. The phytocompounds of the oil under study were dif-
ferent from those of the earlier reports wherein (E)cinnamyl acetate and
P-caryophyllene were the main constituents. The oil has been reported to contain
phenyl propanoids, oxygenated monoterpenes, monoterpenes, and sesquiterpenes.
The main constituents of the oil were a-pinene, P-pinene, P-caryophyllene,
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 657
a-muurolene, y-cadinene, 3-cadinene, and a-muurolol. The oil was devoid of euge-
nol, E-cinnamaldehyde, benzyl benzoate, and camphor which are major constitu-
ents of the leaf, stem bark, and root oils of C. zeylanicum.
Senanayake et al. [78] reported that C. zeylanicum essential oil contained several
resinous compounds, such as cinnamic acid, cinnamaldehyde, and cinnamate. A
spicy flavor and a strong aroma of Cinnamomum were reported due to the presence
of cinnamaldehyde. Trans-cinnamaldehyde, terpinolene, cinnamyl acetate, eugenol,
caryophyllene oxide, L-borneol, b-caryophyllene, E-nerolidol, alpha-cubebene,
L-borneol acetate, alpha-terpineol, and alpha thujene were some of the essential oils
found in C. zeylanicum [63, 79, 80]. Aldehydes, esters, phenols, acids, diterpenes,
sesquiterpenes, monoterpenes, benzopyrones, hydrocarbon alcohols, and flavonoids
were the chemical substances found in C. zeylanicum. Aldehydes present in C. zey-
lanicum bark essential oil were methoxycinnamaldehyde, benzenepropanal, cinna-
maldehyde, vanillin, cuminaldehyde, benzaldehyde, hydrocinnamic,
2-methyl-3-phenyl-propanal, and citronellal. Alcohol groups present in C. zeylani-
cum were cinnamyl alcohol, α-terpineol, linalool, α-bisabolol, cinnamyl acetate
esters, cinnamaldehyde, methyl cinnamate, hydrocinnamyl acetate, benzyl benzo-
ate, and bornyl acetate [47, 81]. Brari and Thakur [74] reported cinnamaldehyde and
linalool from essential oil isolated from C. zeylanicum.
Kamalakannan et al. [82] isolated hymecromone and umbelliferone from etha-
nolic extract of C. cassia. Cinnamomum cassia contains volatile oils with cinnamic
acid, eugenol, cinnamyl alcohol, cinnamaldehyde, melilotic acid, δ-cadinene, phe-
nolic compounds, epicatechins, cinnamic aldehydes, monoterpenes, tannins, procy-
anidins, diterpenes, glycosides (cinnacassides A–Z), oxalate, sesquiterpenes
(pinene), and traces of coumarin [83]. Packiaraj et al. [84] reported major com-
pounds such as NDidehydrohexacarboxyl-2,4,5-trimethylpiperazine,
1,2,4-triazoliumylide phenol, 3,5-dimethoxy acetate, and 4′-isopropylidene-bis-
(2-cyclohexyl) phenol. Coumarin (1,2-benzopyrone) content was reported with a
major difference between C. cassia and C. zeylanicum in their vegetative parts [85].
Tanaka et al. [86] isolated 3-(2-hydroxyphenyl)-propanoic acid and its
O-glucoside from the stem bark of C. cassia. Chemical compounds of C. cassia
were coumarin, (Z)-cinnamaldehyde, α-ylangene, and β-caryophyllene [87–89].
Barks and leaves of C. cassia contain cinnzeylanol, 19-dehydroxy-13-
hydroxycinncassiol, (18R)-1-hydroxycinncassiol, (18S)-3-dehydroxycinncassiol
glucoside, (18S)-3-dehydroxy-8-hydroxycinncassiol, (18S)-cinncassiol, (18S)-3,5-
didehydroxy- 1,8-dihydroxycinncassiol, and 2,3-dihydroxy-1-(4-hydroxy-3,5-
dimethoxyphenyl)-1-propanone [90, 91]. Leaves contain
(1R,2R)-4-[(3S)-3-hydroxybutyl]-3,3,5- trimethylcyclohex-4-ene-1,2-diol,
(3S,5R,6R,7E,9S)-3,5,6,9-tetrahydroxy-7-enemegastigmane, and (1R,2R,4S,6S)-4-
(2-hydroxypropan-2-yl)-1-methyl-7-oxabicyclo[4.1.0]heptan-2-ol dimethanol [90].
The twig of C. cassia was reported to contain certain chemical compounds such
as cinnamyl alcohol and 2-hydroxy-cinnamyl alcohol [61, 92], (+)-syringaresinol,
cinnamomulactone, 2-hydroxycinnamaldehyde [61, 91–93], cinnamic acid [92],
and phenethyl (E)-3-[4-methoxyphenyl]-2-propenoate [61]. Chemical constituents
reported in C. cassia leaves were 1-(3,4-dimethoxyphenyl)-1,2,3-propanetriol [90],
658 S. Surendran and R. Ramasubbu
Nath et al. [32] carried out GC-MS analysis in C. tamala essential oil from Assam,
India. α-Linalool, α-pinene, and pinene were the major constituents, whereas cin-
namaldehyde and eugenol were the minor constituents.
Cinnamomum tamala leaves collected from Dehradun, Uttarakhand, contain cin-
namaldehyde, cis-linalool oxide, linalool, and cinnamyl acetate as the major con-
stituents. Benzaldehyde, 1,8-cineole, bornyl acetate, 3-phenyl propanal, and
p-cymene were the minor constituents [104]. Gulati et al. [105] reported linalool
and cinnamaldehyde from the two samples of C. tamala from the Kumaun region.
Cinnamaldehyde was reported as the main compound of C. tamala (Kubeczka and
Formacek 2002) [106–108]. Cinnamomum tamala oil samples were also reported to
contain cinnamic acid [109]. Showkat et al. [110] identified chemical constituents
such as β-caryophyllene, germacrene A, β-sabinene, α-pinene, myrcene, (Z)-β-
ocimene, linalool, α-gurjunene, and trans-sabinene hydrate in C. tamala leaf essen-
tial oil. Leaf samples were detected with three flavonoid compounds: quercetin,
quercetin, and kaempferol [112]. Eugenol was the principal constituent in C. tamala
essential oil followed by eugenyl acetate and α-phellandrene [55]. Cinnamomum
tamala leaf volatile oil was reported to contain eugenol which is the major constitu-
ent [50, 112].
2,6,10-Trimethyl-12-oxatricyclo[7.3.0.0{1,6}]tridec-2-ene and hexahydropyri-
dine,4- [4,5-dimethoxyphenyl]-in were isolated from hexane extract, and three
compounds from dichloromethane extract, namely, 2,5-chloro-3β-
hydroxy-6βnitro-5α-androstan-17-one, acetic acid,10,13-dimethyl-2-oxo-
2,3,4,7,8,9,10,11,12,13,14,15,16,17-tetradecahydro-1H cyclopenta
[a]phenanthren17-ylester, and 6á,19-CycloAndrost-4-ene-3,17-dione were reported
from extracts of C. tamala [113]. Heer et al. [114] reported 21 compounds from
fresh leaves of C. tamala essential oil collected from northwestern Himalaya by
GC-MS analysis. They consist of complex mixtures of monoterpenes, phenylpro-
panoids, and sesquiterpenes. Kumar et al. [103] reported that C. tamala leaves con-
tain several phytochemicals such as β-caryophyllene, trans-cinnamyl acetate,
eugenol, and cinnamaldehyde. Srivastava et al. [115] reported the GC-MS analysis
of C. camphora oil, and the major constituents were fenchone, camphene, a-thujene,
L-limonene, and cisp-menthane. Camphor was found in C. camphora from
Pantnagar, Uttarakhand [53]
Ghalib et al. [116] reported C. iners from chloroform and alcoholic leaf extracts.
Nine components were detected as major components. Eicosanoic acid ethyl ester
and caryophyllene are the most prominent components of the chloroform extract.
Caryophyllene was the major compound of the alcohol extract. Udayaprakash et al.
[117] reported six compounds, i.e., pentadecanoic acid, 14-methyl-, methyl ester;
4-piperidineacetic acid, 10-octadecenoic acid, methyl ester; cyclopropanebutanoic
acid, 2-[[2-[[2-[(pentylcyclopropyl) methyl] cyclopropyl] methyl] cyclopropyl]
methyl]-, methyl ester; cyclopentaneundecanoic acid, methyl ester; 1-acetyl-5-
ethyl2-[3-(2-hydroxyethyl)-1H-indol-2-yl]-a-methyl-methylester; and 3-pentyl-,
methyl ester, oxiraneundecanoic acid, found in the essential oil of leaves of C. iners
by GC-MS analysis.
660 S. Surendran and R. Ramasubbu
Bullerman et al. [139] reported that the bark oil of C. zeylanicum inhibited fungal
growth and aflatoxin production due to the presence of eugenol and cinnamalde-
hyde. Montes-Belmont and Carvajal [140] reported fungitoxic properties against
fungi involved in respiratory tract mycoses such as Aspergillus niger, A. fumigatus,
A. nidulans, and A. flavus. Simic et al. [39] reported that C. zeylanicum oil has the
strongest antifungal activity due to the presence of trans-cinnamaldehyde as the
major component. A study has been reported that 80% of bacteria and fungi were
killed by cinnamaldehyde [141]. Choudhary et al. [142] reported the antimicrobial
activity of Cinnamomum cassia essential oil against several bacterial cultures.
About 99.4% of the organisms including Streptococcus oralis, Micrococcus roseus,
S. anginosus, S. sanguinis, S. intermedius, and Enterobacter aerogenes were inhib-
ited, but it was not effective against Salmonella Paratyphi B.
Biavati et al. [143] studied the antimicrobial effects of C. cassia aqueous infu-
sion and observed inhibition in the microbial strains such as Micrococcus roseus,
S. intermedius, S. anginosus, S. mutans, S. sanguis, S. oralis, S. morbillorum, S. sal-
ivarius, S. uberis, Klebsiella pneumonia, and Flavobacterium. Rameshkumar et al.
[144] reported that C. filipedicellatum essential oil showed moderate activity against
gram-positive and gram-negative bacteria such as Salmonella Typhi and
Staphylococcus aureus, and no inhibition was observed in Pseudomonas aerugi-
nosa. Dongmo et al. [145] studied the antifungal activity of C. zeylanicum essential
oil from Cameroon against some common fungi causing spoilage of stored food
Table 1 Chemical compounds reported in Cinnamomum spp
Sl. no Plant name Distribution Phytocompounds isolated Source of compound Reference
1. Cinnamomum India [Karnataka, Kerala, Linalool, (E)-caryophyllene (E)-cinnamyl acetate Petiole, terminal Essential oils Leela et al. [28]
malabatrum Tamil Nadu]; endemic bicyclogermacrene, benzyl benzoate, caryophyllene shoot, leaf, and shoot
(Burm.f.) J.Presl oxide, and humulene epoxide II
3,4′,5,7-Tetrahydroxyl flavones, Leaves Extract Agrawal et al. [22]
3,3′,4′,5,7-pentahydroxy flavones, kaempferol-3-O-
sophoroside, and quercetin 3-O-rutin
Eugenol, β-caryophyllene, cinnamic aldehyde, Bark Essential oils Agrawal et al. [22]
benzaldehyde, camphor, cadinene, limonene, geraniol,
ocimene, γ-terpinene, eugenol acetate, benzyl
cinnamate, β-phellandrene, α-terpineol, and benzyl
acetate, cinnamaldehydes
Alkaloids, tannins, glycosides, triterpenoids, flavonoids, Leaf Extract Natarajan et al. [32]
and saponins
Cinnamic aldehyde, benzaldehyde, eugenol, camphor, Leaves Essential oil Agrawal et al. [22]
cadinene, α-terpineol, limonene, geraniol, eugenol
acetate, ocimene, β-caryophyllene, γ-terpinene,
β-phellandrene, benzyl cinnamate, and benzyl acetate
Cinnamaldehydes. kaempferol-3-O-sophoroside, Bark Essential oil Agrawal et al. [22]
3,4′,5,7-tetrahydroxyl flavones, 3,3′,4′,5,7-pentahydroxy
flavones, and quercetin 3-O-rutin
N-Didehydrohexacarboxyl-2,4,5-trimethylpiperazine, Leaf Packiaraj et al. [85]
1,2,4-triazoliumylide phenol 3,5-dimethoxy acetate,
4′-isopropylidene-bis-(2-cyclohexyl) phenol
Cinnamaldehydes, kaempferol-3-O-sophoroside, Bark Essential oil Agrawal et al. [22]
3,4′,5,7-tetrahydroxyl flavones, 3,3′,4′,5,7-pentahydroxy
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff
(continued)
Table 1 (continued)
668
(continued)
673
Table 1 (continued)
674
and one fungal strain by agar well diffusion method. The extract showed variable
degree of inhibition zones except for dichloromethane, aqueous fraction, and crude
extract which were found to be completely inactive against Salmonella Typhi (a
gram-negative strain). Adarsh et al. [173] reported significant antimicrobial activity
of C. zeylanicum against Escherichia coli (gram-negative), Enterococcus faecalis
(gram-positive), and Salmonella Typhi (gram-positive) by agar diffusion method.
Naik et al. [174] assessed the antimicrobial activity of two cinnamon leaf oils and
extracts by disc diffusion assay and the minimum inhibitory concentration by two-
fold serial dilution method against E. coli, S. Typhi, S. aureus, B. cereus, and C. per-
fringens. Essential oils and extracts exhibited the highest zone of inhibition (ZOI)
against S. aureus and E. coli. Both oils and extracts showed minimum inhibitory
concentration in the range of 0.156 mg/ml to 5 mg/ml.
Cong et al. [175] demonstrated the antimicrobial activity of leaf essential oil
from C. longipaniculatum against Staphylococcus aureus, Bacillus subtilis, Sarcina
lutea, and Salmonella typhimurium. Chairunnisa et al. [176] reported that volatile
compounds such as α-pinene, α-terpineol, 1,8-cineole, and trans-cinnamaldehyde
from C. burmannii essential oil exhibit antibacterial activity against Escherichia
coli and Staphylococcus aureus. Aqueous extracts from C. camphora leaves exhibit
positive effect on Penicillium purpurogenum, Trichoderma harzianum, Aspergillus
fumigatus, Phanerochaete chrysosporium, and Gloeophyllum trabeum with concen-
trations of 5% and 10% found to be effective against Botryodiplodia theobromae
(Hu et al. 2017) [177].
Rangel et al. [178] reported the antifungal activity of C. zeylanicum leaf essential
oil against Candida spp. with MIC and MFC values ranging from 62.5 to 1000 μg/
mL. Cinnamomum cassia essential oil was reported to contain cinnamaldehyde,
cinnamic acid, and benzaldehyde as the major constituents and with remarkable
antibacterial activity against Escherichia coli, Staphylococcus hyicus,
Staphylococcus aureus, Propionibacterium acnes, and Pseudomonas aeruginosa
[179–181]. Lu et al. [182] reported that C. cassia acetone extract exhibited antifun-
gal activity against Alternaria alternata, Botrytis cinerea, Colletotrichum glycines,
Fusarium decemcellulare, and Alternaria solani with the half-maximal effective
concentration ranging from 45.68 mg/L to 105.09 mg/L.
Lin et al. [183] evaluated antioxidant activities of aqueous and ethanol extracts from
Cinnamomum cassia dry bark. At a concentration of 1.0 mg/mL, C. cassia ethanol
extracts exhibit greater inhibition than α-tocopherol. The same extract also showed
an excellent antioxidant activity in enzymatic and nonenzymatic liver tissue oxida-
tive systems. Ethanolic extract of C. cassia revealed the strongest antioxidant activ-
ity followed by α-tocopherol. The IC50 values of ethanolic extract of C. cassia
compared to α-tocopherol were found to be lower in thiobarbituric acid test
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 681
analgesic activity, maximum time required for response against thermal stimuli was
observed in ethanol extract and maximum % of writhing inhibition (44.57%) when
compared to aqueous extract. Chua et al. [191] reported the antioxidant activity of
C. osmophloeum ethanolic extracts from the twigs of C. osmophloeum. BuOH frac-
tion exhibited the best performance and consequently, kaempferol-7-O-rhamnoside
was also isolated and its activity was confirmed.
Chakraborty and Das [192] evaluated the antihyperglycemic activity of C. tamala
leaf aqueous extracts. Quantification of antioxidants of the leaves – phenols, ascor-
bate, and carotenoids – revealed that C. tamala leaves had high antioxidants. Anis
et al. [193] investigated the antioxidant activity of extracts from C. iners wood. The
ethanol extract showed EC50 value of 14.96 μg/mL with the highest antioxidant
activity followed by chloroform extract with EC50 > 30 μg/mL. No activity was
observed in water extract. Park et al. [194] evaluated the antioxidant activity of
C. verum extract by supercritical fluid extracts and Marc methanol extracts. Higher
antioxidant activities were observed in DPPH and ABTS radical scavenging assay.
Srinivasa et al. [195] reported that C. aromaticum showed significant antioxidant
activity and was used as a natural antioxidant agent. Abeysekera et al. [196] reported
that C. zeylanicum ethanolic extracts of both leaf and bark had significantly high
antioxidant activity. Abdelwahab et al. [138] reported that C. altissimum bark extract
displayed antioxidant activities with IC50 value of 38.5 ± 4.72 μg/ml using DPPH
assay and 345.2 ± 14.8 (μM Fe (II)/g dry mass) using FRAP assay.
Salleh et al. [197] reported the antioxidant and anticholinesterase activity of
C. griffithii and C. macrocarpum essential oil. The bark oil of C. griffithii exhibits
IC50 value of 73.4 μg/mL on DPPH assay, while the leaf oil showed inhibition
value of 65.5 μg/mL. Cinnamomum macrocarpum bark oil exhibits inhibition val-
ues of 55.8% and 66.1% at 1 mg/mL concentration. Udayaprakash et al. [117] con-
ducted antioxidant studies on C. iners methanolic leaf extract. DPPH free radical
scavenging activity of methanolic leaf extract recorded an IC50 value at the concen-
tration of 15 g/ml. ABTS assay (99.36%) showed maximum inhibition followed by
TBA (95.39%) and FTC (81.37%). Brodowska et al. [198] carried out the antioxi-
dant activity of C. cassia essential oils. Lower IC50 value was observed in DPPH
and ABTS assay (IC50 = 42.03 μg/L and IC50 = 5.13 μg/L) for cinnamon extracts
and indicates higher radical scavenging activity. Extracts were found to be better
radical scavenger than essential oils with IC50 values of 64.51 μg/L (ABTS)
and147.23 μg/L (DPPH).
Valizadeh et al. [170] conducted an antioxidant study on C. zeylanicum barks and
leaves by DPPH assay. Free radical scavenging activity was found to be increased
by increasing C. zeylanicum essential oil concentration. The concentration of CEO
resulting in 50% inhibition of the free radical (IC50) was 79.54 μg/mL. Ervina et al.
[199] reported that the C. zeylanicum bark infusion showed the highest antioxidant
activity with an IC50 value of 3.03 followed by ethanolic extract and its water and
ethyl acetate fractions with IC50 values of 8.36, 8.89, and 13.51 μg/mL, respec-
tively. Fu et al. [200] reported antioxidative effect in diet-induced obese rats by seed
kernel oil of C. camphora. Liu et al. [201] reported that ferric scavenging activity
test on C. longipaniculatum leaves displayed a higher reducing activity of
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 683
proanthocyanidins compared to vitamin C and BHT but lower than BHA by ferric
scavenging activity test. Potassium ferricyanide reduction method confirmed a
higher antioxidant activity than BHA (0.094 mg/mL), vitamin C (0.125 mg/mL),
and BHT (0.125 mg/mL) when the proanthocyanidin concentration was
0.156 mg/mL.
Liu et al. [202] evaluated the antioxidative activity of the flavonoids isolated
from C. camphora leaves. The flavonoids exhibited DPPH free radical scavenging
activity similar to the positive control of vitamin C with increasing concentration.
The reducing ability also increased significantly with the increase of concentration
and was very close to vitamin C, BHA, and BHT. Kallel et al. [203] reported high
cytotoxicity cell line effect in C. zeylanicum essential oil. In vitro cytotoxicity was
examined using an MTT assay against HeLa and Raji cell lines. The essential oil
inhibited the proliferation of HeLa and Raji cell lines and showed IC50 values of
0.57 μg/mL and 0.13 μg/mL. Priani et al. [204] reported that strong antioxidant
activity with IC50 value of 10.04 ± 0.08 ppm was observed in the bark of C. bur-
mannii. A peel-off mask which significantly exhibits potent antioxidant effects
(IC50 = 47.31 ± 1.47 ppm) was formulated. Ribeiro et al. [205] reported the antioxi-
dant activity of leaf and stem of C. zeylanicum by using DPPH method. The inhibi-
tion percentage for the leaves was 59.17 ± 0.11% and for the stem was 61.34 ± 0.11%.
Raksha et al. [206] conducted an antioxidant activity on C. tamala leaf extracts
by DPPH free radical assay and observed significant antioxidant activity. The
hydroalcoholic leaf extract at a 100 μm/ml concentration exhibited inhibition activ-
ity of about 96.99 ± 0.99%. Singh et al. [207] reported the antioxidant and antidia-
betic effect of C. cassia bark methanolic extracts. In acute toxicity testing, up to
2000 mg/kg methanolic extracts did not show any significant toxic signs; hence, the
antidiabetic activity was carried out at 125, 250, and 500 mg/kg dose levels. The
diabetic animals showed significant increases in the levels of total cholesterol (TC),
very-low-density lipoprotein, and TC/high-density lipoprotein radio compared with
that of normal control and also the extracts prevent STZ-induced hyperlipidemia. In
the histopathological analysis, sections from the liver, pancreas, and kidney of the
diabetic animals and the animals treated with MECC 500 mg/kg showed mid-to-
moderate toxic effects.
Kamath et al. [218] evaluated the wound healing activity of C. zeylanicum bark
ethanolic extract in Wistar rats. The extract at doses of 250 mg/kg and 500 mg/kg
body weight significantly enhance the wound breaking strength, of wound contrac-
tion and epithelialization period. In the dead space wound, the granulation tissue
weight, hydroxyproline content, and breaking strength were also increased by the
extract. Soni et al. [219] reported that active extract from ethanolic extract of
C. tamala leaves was responsible for the wound healing activity in diabetic Wistar
albino rats. Both the wound area and day of epithelialization were significantly
decreased in the excision wound model. Significantly higher tensile strength was
observed in the rats treated orally with ethanolic extract treated in incision wound
model. Weights of wet and dry granulation tissue also increase with increased
amounts of hydroxyproline, elastin, and collagen.
Narkhede et al. [220] reported the wound healing activity of C. zeylanicum and
C. tamala in Sprague Dawley rats. The time taken for complete epithelialization and
wound contraction was significantly less than the control. The mean tensile strength
was significantly greater after 16 days. Methanolic extract showed better granula-
tion tissues, better tensile strength, and early and complete epithelialization. Deepa
et al. [221] reported that the hydroalcoholic extract of C. nitidum stem bark showed
dose-dependent percentage wound healing. Significant wound contraction and high
degree of tensile strength were observed in treated animals as compared with the
control. Hydroxyproline level was found to be significantly increased in a dose-
dependent manner.
Ahmadi et al. [222] evaluated the effects of an ointment prepared from C. verum
essential oil in infected wound model. Topical administration of C. verum remark-
ably shortened the inflammatory phase, increased fibroblast distribution and colla-
gen deposition, and accelerated the cellular proliferation, reepithelialization, and
keratin synthesis. The mRNA levels of IGF-1, FGF-2, and VEGF were remarkably
686 S. Surendran and R. Ramasubbu
higher in C. verum-treated groups (especially 2%) than in the control group. Topical
administration of C. verum increased the antioxidant power and reduced the MDA
content in comparison to control animals. C. verum accelerates wound healing by
upregulating the IGF-1, FGF-2, and VEGF expression and increasing cell prolifera-
tion, collagen synthesis, and reepithelialization ratio.
Kefayat et al. [223] reported cinnamon extracts were incorporated into the bacte-
rial cellulose membranes to prepare an all-natural wound dressing. The cinnamon
extract membrane maintains appropriate moisture content for an acceptable period
of time. Although the tensile strength and elongation at break values of the cinna-
mon extract were slightly lower than the BC membrane, they are still in ideal ranges.
The cinnamon extract membrane exhibits significantly more antibacterial effects
against Staphylococcus aureus and Escherichia coli, and they are also found to be
more biocompatible with L929 normal skin fibroblast cells than with the bacterial
cellulose and chitosan membranes.
4 Conclusion
References
1. Pitman NC, Jørgensen PM (2002) Estimating the size of the world’s threatened flora. Science
298(5595):989–989
2. Veeresham C (2012) Natural products derived from plants as a source of drugs. J Adv Pharm
Technol 3(4):200
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 687
3. Seth SD, Sharma B (2004) Medicinal plants in India. Indian J Med Res 120(1):9
4. Fabricant DS, Farnsworth NR (2001) The value of plants used in traditional medicine for
drug discovery. Environ Health Perspect 109(suppl 1):69–75
5. Newman DJ, Cragg GM (2007) Natural products as sources of new drugs over the last 25
years. J Nat Prod 70(3):461–477
6. Kharwar RN, Maurya AL, Verma VC et al (2012) Diversity and antimicrobial activity of
endophytic fungal community isolated from medicinal plant Cinnamomum camphora. Proc
Natl Acad Sci India Sect B Biol Sci 82(4):557–565
7. Schmidt E, Jirovetz L, Buchbauer G et al (2006) Composition and antioxidant activities of
the essential oil of cinnamon (Cinnamomum zeylanicum Blume) leaves from Sri Lanka. J
Essent Oil-Bear Plants 9(2):170–182
8. Maridass M, Victor B (2008) Ethnobotanical uses of Cinnamomum species, Tamil Nadu,
India. Ethnobot Leafl 2008(1):18
9. Burkill H (1985) The useful plants of West Tropical Africa. Royal Botanic Garden
Kew, London
10. Wagner H, Fessler B, Geyer B et al (1986) 5-lipoxygenase-inhibitors from medicinal plants.
Planta Med 52(06):549–550
11. Mustaffa F, Indurkar J, Shah M et al (2013) Review on pharmacological activities of
Cinnamomum iners Reinw. ex Blume. Nat Prod Res 27(10):888–895
12. Pengelly A (2004) Constituents of medicinal plants. CABI Publishing, Cambridge, p 66
13. Maridass M (2008) Anti-inflammatory activity of the methanolic extract of Cinnamomum
sulphuratum barks. Ethnobot Leafl 2008(1):63
14. Maridass M (2009) Screening of antifungal activities of barks of Cinnamomum species. Thai
J Pharm Sci 33:137–143
15. Rameshkumar KB, George V (2006) Cinnamomum sulphuratum Nees – a benzyl benzoate-
rich new chemotype from southern western ghats, India. J Essent Oil Res 18(5):521–522
16. Shivaprasad D (2015) Reproductive biology of Cinnamomum sulphuratum Nees. From wet
evergreen forest of Western Ghats in Karnataka. Proc Int Acad Ecol Environ Sci 5(1):7
17. Lemonica IP, Macedo AB (1994) Abortive and/or embryofetotoxic effect of Cinnamomum
zeylanicum leaf extracts in pregnant rats. Fitoterapia 65(5):431–434
18. Kubo M, Ma S, Wu J, Matsuda H (1996) Anti-inflammatory activities of 70% methanolic
extract from Cinnamomi Cortex. Biol Pharm Bull 19(8):1041–1045
19. Zhang C, Fan L, Fan S et al (2019) Cinnamomum cassia Presl: a review of its traditional uses,
phytochemistry, pharmacology and toxicology. Molecules 24(19):3473
20. Annegowda HV, Gooi TS, Awang SHH et al (2012) Evaluation of analgesic and antioxidant
potency of various. Int J Pharmacol 8(3):198–203
21. Kumar BH, Basheer S (2010) Antioxidant potential and antimicrobial activity of Cinnamomum
Malabathrum (Batka). Orient J Chem 26(4):1449
22. Agarwal SK, Chipa RC, Samantha Suresh KC (2013) Anticancer activity of Cinnamomum
Malabatrum against Dalton’s ascitic lymphoma. Int J Res Pharmacol Pharmacother
2:314–319
23. Kurokawa M, Kumeda CA, Yamamura J et al (1998) Antipyretic activity of cinnamyl deriva-
tives and related compounds in influenza virus-infected mice. Eur J Pharmacol 348(1):45–51
24. Kar A, Choudhary BK, Bandyopadhyay NG (1999) Preliminary studies on the inorganic
constituents of some indigenous hypoglycaemic herbs on oral glucose tolerance test. J
Ethnopharmacol 64(2):179–184
25. Hasler CM, Blumberg JB (1999) Phytochemicals: biochemistry and physiology. Introduction.
J Nutr 129(3):756S–757S
26. Costa MA, Zia ZQ, Davin LB, Lewis NG (1999) Chapter four: toward engineering the meta-
bolic pathways of cancer- preventing lignans in cereal grains and other crops. In: Romeo JT
(ed) Recent advances in phytochemistry, phytochemicals in human health protection, nutri-
tion, and plant defense, vol 33. Kluwer Academic/Plenum Publishers, New York, pp 67–87
27. Porrini M, Riso P (2008) Factors influencing the bioavailability of antioxidants in foods: a
critical appraisal. Nutr Metab Cardiovasc 18(10):647–650
688 S. Surendran and R. Ramasubbu
28. Leela NK, Vipin TM, Shafeekh KM et al (2009) Chemical composition of essential oils
from aerial parts of Cinnamomum malabatrum (Burman f.) & Presl. Bercht. Flavour Fragr J
24(1):13–16
29. Aravind R, Bindu AR, Bindu K, Alexeyena V (2014) GC-MS analysis of the bark essential
oil of Cinnamomum malabatrum (Burman. F) Blume. Res J Pharm Technol 7(7):754–759
30. Anil AM, Bency BJ, Helen PM, Rani DS (2018) Docking and in vitro studies on antioxidant,
antibacterial and cytotoxic properties of cinnamon (Cinnamomum malabathrum). Inter J Res
Analy Rev 5(5):66–72
31. Natarajan P, John S, Thangatirupati A, Kala R (2014) Antihyperlipidemic activity of alco-
holic extract of Cinnamomum malabatrum burm. On cholesterol diet induced rats. World J
Pharm Res 3(6):1599–1615
32. Nath SC, Hazarika AK, Baruah RN et al (1994) Major components of the leaf oil of
Cinnamomum sulphuratum Nees. J Essent Oil Res 6(1):77–78
33. Baruah A, Nath SC, Leclercq PA (1999) Leaf and stem bark oils of Cinnamomum sulphura-
tum Nees from Northeast India. J Essent Oil Res 11(2):194–196
34. Baruah A, Nath SC, Hazarika AK (2002) Essential oils of Cinnamomum sulphuratum Nees-A
new chemotype source of spice value from Northeast India. Indian Perfum 46:89–92
35. Baruah A, Nath SC, Hazarika AK (2001) Methyl cinnamate, the major component of the leaf
and stem bark oils of Cinnamomum sulphuratum Nees. Indian Perfum 45:39–41
36. Kumar KNS, Rajalekshmi M, Sangeetha B et al (2013) Chemical fingerprint of leaves of
Cinnamomum sulphuratum Nees growing in Kodagu, Karnataka. J Pharmacogn Phytochem
2:163–168
37. Singh C, Singh S, Pande C et al (2014) Chemical composition of the leaves essential oil
from Cinnamomum glanduliferum (Wall) Meissn from Uttarakhand, India. J Essent Oil Bear
Plants 17(5):927–930
38. Rao BR, Rajput DK, Bhattacharya AK (2007) Essential oil composition of petiole of
Cinnamomum verum Bercht. & Presl. J Spices Aromat Crops 16:38–41
39. Simic A, Sokovic MD, Ristic M et al (2004) The chemical composition of some Lauraceae
essential oils and their antifungal activities. Phytother Res 18:713–717
40. Mollenbeck S, Konig T, Schreier P et al (1997) Chemical composition and analysis of enan-
tiomers of essential oils from Madagascar. Flavour Fragr J 12:63–69
41. Variyar PS, Bandyopadhyay C (1989) On some chemical aspects of Cinnamomum zeylani-
cum. Pafai J 10(4):35–38
42. Mallavarapu GR, Ramesh S, Chandrasekhara RS et al (1995) Investigation of the essential oil
of cinnamon leaf grown at Bangalore and Hyderabad. Flavour Fragr J 10:239–242
43. Rao BRR, Rajput DK, Kaul PN et al (2006) Effect of short and long-term storage on essential
oil content and composition of cinnamon (Cinnamomum verum Bercht. & Presl.). J Spices
Aromat Crops 15:19–24
44. Nath SC, Pathak MG, Baruah A (1996) Benzyl benzoate, the major component of the leaf and
stem bark oil of Cinnamomum zeylanicum Blume. J Essent Oil Res 8:327–328
45. Wijesekera ROB, Jayawardene AL, Rajapakse LS (1974) Volatile constituents of leaf, stem
and root oils of cinnamon (Cinnamomum zeylanicum). J Sci Food Agric 25:1211–1220
46. Kaul PN, Bhattacharya AK, Rajeswara Rao BR et al (2003) Volatile constituents of essential
oils isolated from different parts of cinnamon (Cinnamomum zeylanicum Blume). J Sci Food
Agric 83:53–55
47. Jayaprakasha GK, Jaganmohan Rao L, Sakariah KK (1997) Chemical composition of the
volatile oil from the fruits of Cinnamomum zeylanicum Blume. Flavour Fragr J 12:331–333
48. Mariappan PM, Sabesan G, Koilpillai B et al (2013) Chemical characterisation and antifun-
gal activity of methanolic extract of Cinnamomum verum J. Presl bark against Malassezia
spp. Pharmacogn J 5(5):197–204
49. Mir SR, Ali M, Kapoor R (2004) Chemical composition of essential oil of Cinnamomum
tamala Nees et Eberm. leaves. Flavour Fragr J 19(2):112–114
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 689
50. Kapoor IPS, Singh B, Singh G, Isidorov V, Szczepaniak L (2009) Chemistry, antimicrobial
and antioxidant potentials of Cinnamomum tamala Nees & Eberm.(Tejpat) essential oil and
oleoresins. Nat Prod Radiance 8(2):106–116
51. Joshi SC, Padalia RC, Bisht DS, Mathela CS (2009) Terpenoid diversity in the leaf essential
oils of Himalayan Lauraceae species. Chem Biodivers 6(9):1364–1373
52. Chanotiya CS, Yadav A (2010) Enantioenriched (3S)-(+)-linalool in the leaf oil of
Cinnamomum tamala Nees et Eberm. from Kumaon. J Essent Oil Res 22(6):593–596
53. Agarwal R, Pant AK, Prakash O (2012) Chemical composition and biological activities of
essential oils of Cinnamomum tamala, Cinnamomum zeylenicum and Cinnamomum cam-
phora growing in Uttarakhand. In: Chemistry of phytopotentials: health, energy and environ-
mental perspectives. Springer, Berlin/Heidelberg, pp 87–92
54. Kumar KN, Sangeetha B, Rajalekshmi M et al (2012) Chemoprofile of tvakpatra; leaves of
Cinnamomum verum JS Presl. Pharm J 4(34):26–30
55. Rana VS, Langoljam RD, Verdeguer M, Blázquez MA (2012) Chemical variability in the
essential oil of Cinnamomum tamala L. leaves from India. Nat Prod Res 26(14):1355–1357
56. Lohani H, Andola HC, Chauhan N et al (2012) Variability in volatile constituents of
Cinnamomum tamala leaf from Uttarakhand Himalaya. Asian Pac J Trop Biomed
2(2):S667–S669
57. Sankaran V, Chakraborty A, Jeyaprakash K et al (2015) Chemical analysis of leaf essential
oil of Cinnamomum tamala from Arunachal Pradesh, India. J Chem Pharm Sci 8(2):246–248
58. Williams AR, Ramsay A, Hansen TV, Ropiak HM (2015) Anthelmintic activity of trans-
cinnamaldehyde and A-and B-type proanthocyanidins derived from cinnamon (Cinnamomum
verum). Sci Rep 5(1):1–2
59. Yan YM, Fang P, Yang MT et al (2015) Anti-diabetic nephropathy compounds from
Cinnamomum cassia. J Ethnopharmacol 165:141–147
60. Liu YH, Tsai KD, Yang SM et al (2017) Cinnamomum verum ingredient
2-methoxycinnamaldehyde: a new antiproliferative drug targeting topoisomerase I and II in
human lung squamous cell carcinoma NCI-H520 cells. Eur J Cancer Prev 26(4):314–323
61. Kim GJ, Lee JY, Choi HG (2017) Cinnamomulactone, a new butyrolactone from the twigs
of Cinnamomum cassia and its inhibitory activity of matrix metalloproteinases. Arch Pharm
Res 40(3):304–310
62. Alva PP, Suresh S, Nanjappa DP et al (2021) Isolation and identification of quorum sens-
ing antagonist from Cinnamomum verum leaves against Pseudomonas aeruginosa. Life Sci
267:118878
63. Singh G, Maurya S, DeLampasona MP et al (2007) A comparison of chemical, antioxidant
and antimicrobial studies of cinnamon leaf and bark volatile oils, oleoresins and their con-
stituents. Food Chem Toxicol 45(9):1650–1661
64. Raina VK, Srivastava SK, Aggarwal KK et al (2001) Essential oil composition of Cinnamomum
zeylanicum Blume leaves from Little Andaman, India. Flavour Fragr J 16(5):374–376
65. Reynolds JEF (1993) Martindale, the extra pharmacopoeia, 13th edn. Info Access &
Distribution, Singapore
66. Jantan IB, Karim Moharam BA, Santhanam J, Jamal JA (2008) Correlation between chemi-
cal composition and antifungal activity of the essential oils of eight Cinnamomum. Species.
Pharm Biol 46(6):406–412
67. Duke JA (1985) CRC handbook of medicinal herbs. CRC Press, Boca Raton, p 677
68. Uma B, Prabhakar K, Rajendran S, Lakshmi Sarayu Y (2009) Studies on GC/MS spectro-
scopic analysis of some bioactive antimicrobial compounds from Cinnamomum zeylanicum.
J Med Plant Res 8(31):125–131
69. Gupta C, Garg AP, Uniyal RC et al (2008) Comparative analysis of the antimicrobial activity
of cinnamon oil and cinnamon extract on some food-borne microbes. Afr J Microbiol Res
2(9):247–251
70. Vangalapati M, Satya NS, Prakash DS et al (2012) A review on pharmacological activities
and clinical effects of cinnamon species. Res J Pharm Bio Chem Sci 3(1):653–663
690 S. Surendran and R. Ramasubbu
71. Jakhetia V, Patel R, Khatri P et al (2010) Cinnamon: a pharmacological review. J Adv Sci
Res 1(2):19–23
72. Ramos-Nino ME, Clifford MN, Adams MR (1996) Quantitative structure activity relation-
ship for the effect of benzoic acids, cinnamic acids and benzaldehydes on Listeria monocyto-
genes. J Appl Bacteriol 80(3):303–310
73. Gupta C, Garg AP, Uniyal RC, Kumari A (2008) Comparative analysis of the antimicrobial
activity of cinnamon oil and cinnamon extract on some food-borne microbes. Afr J Microbiol
Res 2(9):247–251
74. Brari J, Thakur DR (2015) Insecticidal efficacy of essential oil from Cinnamomum zeyl-
anicum Blume and its two major constituents against Callosobruchus maculatus (F.) and
Sitophilus oryzae (L.). J Agric Technol 11(6):1323–1336
75. Baratta MT, Dorman HJ, Deans SG et al (1998) Antimicrobial and antioxidant properties of
some commercial essential oils. Flavour Fragr J 13:235–244
76. Pragadheesh VS, Saroj A, Yadav A et al (2013) Chemical characterization and antifungal
activity of Cinnamomum camphora essential oil. Ind Crop Prod 49:628–633
77. Mallavarapu GR, Ramesh S (2000) Essential oil of the fruits of Cinnamomum zeylanicum
Blume. J Essent Oil Res 12(5):628–630
78. Senanayake UM, Lee TH, Wills RB (1978) Volatile constituents of cinnamon (Cinnamomum
zeylanicum) oils. J Agric Food Chem 26(4):822–824
79. Malsawmtluangi L, Nautiyal BP, Hazarika T et al (2016) Essential oil composition of bark
and leaves of Cinammoum verum Bertch & Presl from Mizoram, North East India. J Essent
Oil Res 28(6):551–556. https://doi.org/10.1080/10412905.2016.1167131
80. Jayaprakasha GK, Jagan Mohan Rao L et al (2003) Volatile constituents from Cinnamomum
zeylanicum fruit stalks and their antioxidant activities. J Agric Food Chem 51(15):4344–4348
81. Jayaprakasha GK, Rao LJ, Sakariah KK (2002) Chemical composition of volatile oil from
Cinnamomum zeylanicum buds. Z Naturforsch C 57(11–12):990–993
82. Kamalakannan K, Rayar A, Megala L (2016) Isolation of phytochemicals from the bark of
Cinnamomum cassia and antidiabetic study in alloxan induced diabetic rats. World J Pharm
Sci 4:143–149. ISSN (Print): 2321-3310; ISSN (Online): 2321-3086
83. Liao SG, Yuan T, Zhang C et al (2009) Cinnacassides A–E, five geranylphenylacetate glyco-
sides from Cinnamomum cassia. Tetrahedron 65(4):883–887
84. Packiaraj R, Jeyakumar S, Ayyappan N et al (2016) Antimicrobial and cytotoxic activities of
endophytic fungus Colletotrichum gloeosporioides isolated from endemic tree Cinnamomum
malabatrum. Stud Fungi 1(1):104–113
85. Archer AW (1988) Determination of cinnamaldehyde, coumarin and cinnamyl alcohol in cin-
namon and cassia by high-performance liquid chromatography. J Chromatogr A 447:272–276
86. Tanaka S, Yoon YH, Fukui H et al (1989) Antiulcerogenic compounds isolated from Chinese
cinnamon. Planta Med 55(03):245–248
87. Chericoni S, Prieto JM, Iacopini P et al (2005) In vitro activity of the essential oil of
Cinnamomum zeylanicum and eugenol in peroxynitrite-induced oxidative processes. J Agric
Food Chem 53(12):4762–4765
88. Mbaveng AT, Kuete V (2017) Cinnamon species. In: Medicinal spices and vegetables from
Africa. Academic Press, Amsterdam, pp 385–395
89. Bansode VJ (2012) A review on pharmacological activities of Cinnamomum cassia Blume.
Int J Green Pharm 6:102–108
90. Zhou L (2016) Studies on the chemical constituents and immunomodulatory activities of the
leaves of Cinnamomum cassia. Huazhong University of Science and Technology, Wuhan
91. Yang B, Liu S, Liu Y et al (2017) PAHs uptake and translocation in Cinnamomum camphora
leaves from Shanghai, China. Sci Total Environ 574:358–368
92. Chen BJ (2015) Research on chemical constituents of Cinnamomum cassia Presl and
Cinnamomum porrectum (Roxb.) Kosterm. M. S. dissertation, Shandong University of
Traditional Chinese Medicine
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 691
93. He S, Zeng KW, Jiang Y, Tu PF (2016) Nitric oxide inhibitory constituents from the barks of
Cinnamomum cassia. Fitoterapia 112:153–160
94. Zeng JF, Zhu HC, Lu JW et al (2017) Two new geranylphenylacetate glycosides from the
barks of Cinnamomum cassia. Nat Prod Res 31(15):1812–1818
95. Liu X, Yang J, Fu J, Xie TG et al (2018) Phytochemical and chemotaxonomic studies on the
twigs of Cinnamomum cassia (Lauraceae). Biochem Syst Ecol 81:45–48
96. Li Z, Cai Z, Qian S, Chen M (2017) A new lactone from the twigs of Cinnamomum cassia.
Chem Nat Compd 53(2):234–236
97. Guo RL, Yin DZ, Zhou HF et al (2018) Chemical constituents from the immature buds of
Cinnamomum cassia (Lauraceae). Biochem Syst Ecol 78:102
98. Feng D (2016) Research on the chemical constituents of cinnamomum camphora. M. S. dis-
sertation, Shandong University of Traditional Chinese Medicine
99. Xu YW (2016) Research on chemical constituents of Cinnamomum chartophyllum H. W. Li.
M. S. dissertation, Shandong University of Traditional Chinese Medicine
100. Chowdhury AR (1999) Essential oil from Cinnamomum glanduliferum (Wall) Nees. Indian
Perfum 43(2):64–66
101. Baruah A, Nath SC (2006) Leaf essential oils of Cinnamomum glanduliferum (Wall) Meissn
and Cinnamomum glaucescens (Nees) Meissn. J Essent Oil Res 18(2):200–202
102. Prakash B, Singh P, Yadav S et al (2013) Safety profile assessment and efficacy of chemically
characterized Cinnamomum glaucescens essential oil against storage fungi, insect, aflatoxin
secretion and as antioxidant. Food Chem Toxicol 53:160–167
103. Kumar S, Vasudeva N, Sharma S (2012) GC-MS analysis and screening of antidiabetic, anti-
oxidant and hypolipidemic potential of Cinnamomum tamala oil in streptozotocin induced
diabetes mellitus in rats. Cardiovasc Diabetol 11(1):1–11
104. Mohan M, Haider SZ, Sharma A et al (2012) Antimicrobial activity and composition of the
volatiles of Cinnamomum tamala Nees. and Murraya koenigii (L.) Spreng. from Uttarakhand
(India). Asia Pac J Trop Dis 2:S324–S327
105. Gulati BC, Agarwal SG, Thappa RK et al (1977) Essential oil of Tejpat (Kumaon) from
Cinnamomum tamala. Indian Perfum 21:15–20
106. Kubeczka KH, Formacek V (2002) Essential oils analysis by capillary gas chromatography
and carbon-13 NMR spectroscopy, 2nd edn. John Wiley & Sons, New York
107. Dighe VV, Gursale AA, Sane RT et al (2005) Quantitative determination of eugenol from
Cinnamomum tamala nees and eberm. Leaf powder and polyherbal formulation using reverse
phase liquid chromatography. Chromatographia 61(9):443–446
108. Seth R, Mohan M, Singh P et al (2012) Chemical composition and antibacterial properties of
the essential oil and extracts of Lantana camara Linn. from Uttarakhand (India). Asian Pac J
Trop Biomed 2(3):S1407–S1411
109. Husain A, Virmani OP, Sharma A et al (1988) Major essential oil bearing plants of India.
Central Institute of Medicinal and Aromatic Plants (CIMAP), CSIR, Lucknow
110. Showkat RM, Ali M, Kapoor R (2004) Chemical composition of essential oil of Cinnamomum
tamala Nees & Eberm leaves. Flavour Fragr J.19:112–114
111. Prasad NK, Yang B, Dong X et al (2009) Flavonoid contents and antioxidant activities from
Cinnamomum species. Innov Food Sci Emerg Technol 10:627–632
112. Dighe VV, Gursale AA, Charegaonkar GA (2009) Quantitation of Eugenol, Cinnamaldehyde
and Isoeugenol from Cinnamomum tamala Nees and Eberm. leaf powder and Cinnamomum
zeylanicum Breyn stem bark powder by LC. Chromatographia 70(11):1759–1762
113. Khajapeer KV, Krishna PP, Baskaran RA (2015) GC MS and elemental analysis of
Cinnamomum tamala. Int J Pharm Pharm Sci 7:398–402
114. Heer A, Guleria S, Razdan VK (2017) Chemical composition, antioxidant and antimicrobial
activities and characterization of bioactive compounds from essential oil of Cinnamomum
tamala grown in north-western Himalaya. J Plant Biochem Biotechnol 26(2):191–198
692 S. Surendran and R. Ramasubbu
115. Srivastava B, Singh P, Shukla R, Dubey NK (2008) A novel combination of the essential oils
of Cinnamomum camphora and Alpinia galanga in checking aflatoxin B1 production by a
toxigenic strain of Aspergillus flavus. World J Microbiol Biotechnol 24(5):693–697
116. Ghalib RM, Hashim R, Sulaiman O et al (2012) Phytochemical analysis, cytotoxic activ-
ity and constituents–activity relationships of the leaves of Cinnamomum iners (Reinw. ex
Blume-Lauraceae). Nat Prod Res 26(22):2155–2158
117. Udayaprakash NK, Ranjithkumar M, Deepa S et al (2015) Antioxidant, free radical scav-
enging and GC–MS composition of Cinnamomum iners Reinw. ex Blume. Ind Crop Prod
69:175–179
118. Baskaran X, Ebbie MG (2008) Essential oil compounds and antibacterial activity of leaves
of Cinnamomum chemungianum Mohan et Henry (Lauraceae). Ethnobot Leafl 12:565–569
119. Rameshkumar KB, George V, Shiburaj S (2007) Chemical constituents and antibacterial
activity of the leaf oil of Cinnamomum chemungianum Mohan et Henry. J Essent Oil Res
19(1):98–100
120. Sriramavaratharajan V, Murugan R (2018) Cumin scented leaf essential oil of Cinnamomum
chemungianum: compositions and there in vitro antioxidant, α-amylase, α-glucosidase and
lipase inhibitory activities. Nat Prod Res 32(17):2081–2084
121. Chen CY, Wang HM, Chung SH et al (2010) Chemical constituents from the roots of
Cinnamomum subavenium. Chem Nat Compd 46(3):474–477
122. Lai Y, Liu T, Sa R et al (2015) Neolignans with a rare 2-oxaspiro [4.5] deca-6, 9-dien-8-one
motif from the stem bark of Cinnamomum subavenium. J Nat Prod 78(7):1740–1744
123. Huang GC, Kao CL, Yeh HC et al (2018) A new diphenyl ether from Cinnamomum subave-
nium. Chem Nat Compd 54(5):869–871
124. Hao X, Sun W, Ke C et al (2019) Anti-inflammatory activities of leaf oil from Cinnamomum
subavenium in vitro and in vivo. Biomed Res Int 2019:1
125. Bakar A, Yao PC, Ningrum V et al (2020) Beneficial biological activities of Cinnamomum
osmophloeum and its potential use in the alleviation of oral mucositis: a systematic review.
Biomedicine 8(1):3
126. Rao PV, Gan SH (2014) Cinnamon: a multifaceted medicinal plant. Evid Based Complement
Alternat Med 2014:1
127. Utchariyakiat I, Surassmo S, Jaturanpinyo M et al (2016) Efficacy of cinnamon bark oil
and cinnamaldehyde on anti-multidrug resistant Pseudomonas aeruginosa and the synergis-
tic effects in combination with other antimicrobial agents. BMC Complement Altern Med
16(1):1–7
128. Jantan IB, Ayop N, Hiong AB, Ahmad AS (2002) Chemical composition of the essential oils
of Cinnamomum cordatum Kosterm. Flavour Fragr J 17(3):212–214
129. Nath SC, Baruah AK (1994) Eugenol as the major component of the leaf oils of Cinnamomum
impressinervium Meissn. J Essent Oil Res 6(2):211–212
130. Baruah A, Nath SC (2007) Cinnamomum champokianum sp. nov. (Lauraceae) from Assam,
Northeastern India. Nord J Bot 25:281–285
131. Hrideek TK, Ginu J, Raghu AV et al (2016) Phytochemical profiling of bark and leaf volatile
oil of two wild Cinnamomum species from evergreen forests of Western Ghats. Plant Arch
16:266–274
132. Sriramavaratharajan V, Stephan J, Sudha V et al (2016) Leaf essential oil of Cinnamomum
agasthyamalayanum from the Western Ghats, India – a new source of camphor. Ind Crop
Prod 86:259–261
133. Sriramavaratharajan V, Sudha V, Murugan R (2016) Characterization of the leaf essential oils
of an endemic species Cinnamomum perrottetii from Western Ghats, India. Nat Prod Res
30(9):1085–1087
134. Cheng SS, Liu JY, Tsai KH et al (2004) Chemical composition and mosquito larvicidal activ-
ity of essential oils from leaves of different Cinnamomum osmophloeum provenances. J Agric
Food Chem 52(14):4395–4400
Phytochemistry and Pharmacological Studies of Indian Cinnamomum Schaeff 693
135. Baruah A, Nath CS (2002) Panicle and bark oils of a variant of Cinnamomum beholghota
(Buch.-Ham.) Sweet from North East India. J Spices Aromat Crop 11(2):135–137
136. Abdelwahab SI, Mariod AA, Taha MME (2017) Chemical composition and antioxidant prop-
erties of the essential oil of Cinnamomum altissimum Kosterm (Lauraceae). Arab J Chem
10(1):131–135
137. Chalchat JC, Valade I (2000) Chemical composition of leaf oils of Cinnamomum from
Madagascar: C. zeylanicum Blume, C. camphora L., C. fragrans Baillon and C. angustifo-
lium. J Essent Oil Res 12(5):537–540
138. Maridass M, Ghanthikumar S (2008) Anti-inflammatory activity of Cinnamomum keralaense
bark extract. Pharmacologyonline 3:322–326
139. Bullerman LB, Lieu FY, Seier SA (1977) Inhibition of growth and aflatoxin production by
cinnamon and clove oils. Cinnamic aldehyde and eugenol. J Food Sci 42(4):1107–1109
140. Montes-Belmont R, Carvajal M (1998) Control of Aspergillus flavus in maize with plant
essential oils and their components. J Food Prot 61(5):616–619
141. McCann J (2003) Herbal Medicine Handbook, 2nd edn. Lippincott, Philadelphia
142. Chaudhry NMA, Tariq P (2006) Anti-microbial activity of Cinnamomum cassia against
diverse microbial flora with its nutritional and medicinal impacts. Pak J Bot 38(1):169
143. Biavati B, Franzoni S, Ghazvinizadeh H (1997) Antimicrobial and antioxidant properties of
plant essential oils. Essential oils. Basic and applied research. In: Proceedings of the 27th
international symposium on essential oils, Vienna, 8–3 September, pp 326–331
144. Rameshkumar KB, George V, Shiburaj S (2006) Chemical constituents and antimicro-
bial activity of the leaf oil of Cinnamomum filipedicellatum Kosterm. J Essent Oil Res
18(2):234–236
145. Dongmo PMJ, Tatsadjieu LN, Tchoumbougnang F et al (2007) Chemical composition, anti-
radical and antifungal activities of essential oil of the leaves of Cinnamomum zeylanicum
Blume from Cameroon. Nat Prod Commun 2(12):1934578X0700201219
146. Ranasinghe L, Jayawardena B, Abeywickrama K (2002) Fungicidal activity of essential
oils of Cinnamomum zeylanicum (L.) and Syzygium aromaticum (L.) Merr et LM Perry
against crown rot and anthracnose pathogens isolated from banana. Lett Appl Microbiol
35(3):208–211
147. Shan B, Cai YZ, Brooks JD, Corke H (2007) The in vitro antibacterial activity of dietary spice
and medicinal herb extracts. Int J Food Microbiol 117(1):112–119
148. Gende LB, Floris I, Fritz R et al (2008) Antimicrobial activity of cinnamon (Cinnamomum
zeylanicum) essential oil and its main components against Paenibacillus larvae from
Argentine. Bull Insectology 61(1):1
149. Gupta C, Garg AP, Uniyal RC et al (2008) Antimicrobial activity of some herbal oils against
common food-borne pathogens. Afr J Microbiol Res 2(10):258–261
150. Aneja KR, Joshi R, Sharma C (2009) Antimicrobial activity of Dalchini (Cinnamomum zeyl-
anicum bark) extracts on some dental caries pathogens. J Pharm Res 2(9):1387–1390
151. Goyal P, Chauhan A, Kaushik P (2009) Laboratory evaluation of crude extracts of
Cinnamomum tamala for potential antibacterial activity. Electron J Biol 5(4):75–79
152. Mishra AK, Mishra A, Kehri HK (2008) Inhibitory activity of Indian spice plant Cinnamomum
zeylanicum extracts against Alternaria solani and Curvularia lunata, the pathogenic dematia-
ceous moulds. Ann Clin Microbiol Antimicrob 8(1):1–7
153. Abdelwahab SI, Zaman FQ, Mariod AA (2010) Chemical composition, antioxidant and anti-
bacterial properties of the essential oils of Etlingera elatior and Cinnamomum pubescens
Kochummen. J Sci Food Agric 90(15):2682–2688
154. Friedman M, Kozukue N, Harden LA (2000) Cinnamaldehyde content in foods determined
by gas chromatography− mass spectrometry. J Agric Food Chem 48(11):5702–5709
155. Bouhdid S, Abrini J, Amensour M (2010) Functional and ultrastructural changes in
Pseudomonas aeruginosa and Staphylococcus aureus cells induced by Cinnamomum verum
essential oil. J Appl Microbiol 109(4):1139–1149
694 S. Surendran and R. Ramasubbu
177. Hu SH, Liu JL, Xu GQ (2017) Inhibitory activity of Cinnamomum camphora leaves extracts
on selected wood fungi and moulds. Forest Sci Technol 76–79
178. Rangel MDL, Aquino SGD, Lima JMD et al (2018) In vitro effect of Cinnamomum zeylani-
cum Blume essential oil on Candida spp. involved in oral infections. Evid Based Complement
Alternat Med 2018:1–13
179. Vaillancourt K, LeBel G, Yi L et al (2018) In vitro antibacterial activity of plant essential oils
against Staphylococcus hyicus and Staphylococcus aureus, the causative agents of exudative
epidermitis in pigs. Arch Microbiol 200(7):1001–1007
180. Li YR, Fu CS, Yang WJ et al (2018) Investigation of constituents from Cinnamomum cam-
phora (L.) J. Presl and evaluation of their anti-inflammatory properties in lipopolysaccharide-
stimulated RAW 264.7 macrophages. J Ethnopharmacol 221:37–47
181. Song SY, Song SH, Bae MS et al (2019) Phytochemical constituents and the evaluation bio-
logical effect of Cinnamomum yabunikkei H. Ohba leaf. Molecules 24(1):81
182. Lu K, Wang QR, Huo X et al (2019) Composition analysis of acetone extract of Cinnamomum
cassia and its inhibition on 5 plant pathogens, Southwest China. J Agric Sci 32:798–802
183. Lin CC, Wu SJ, Chang CH et al (2003) Antioxidant activity of Cinnamomum cassia.
Phytother Res 17(7):726–730
184. Mathew S, Abraham TE (2006) In vitro antioxidant activity and scavenging effects of
Cinnamomum verum leaf extract assayed by different methodologies. Food Chem Toxicol
44(2):198–206
185. Jayaprakasha GK, Negi PS, Jena BS et al (2007) Antioxidant and antimutagenic activities of
Cinnamomum zeylanicum fruit extracts. J Food Compos Anal 20(3–4):330–336
186. Mancini-Filho J, Van-Koiij A, Mancini DA et al (1998) Antioxidant activity of cinnamon
(Cinnamomum Zeylanicum, Breyne) extracts. Boll Chim Farm 137(11):443–447
187. Okawa M, Kinjo J, Nohara T et al (2001) DPPH (1,1-diphenyl-2-picrylhydrazyl) radical
scavenging activity of flavonoids obtained from some medicinal plants. Biol Pharm Bull
24:1202–1205
188. Yang CH, Yang CS, Hwang ML et al (2012) Antimicrobial activity of various parts
of Cinnamomum cassia extracted with different extraction methods. J Food Biochem
36(6):690–698
189. Hsu FL, Li WH, Yu CW et al (2012) In vivo antioxidant activities of essential oils and their
constituents from leaves of the Taiwanese Cinnamomum osmophloeum. J Agric Food Chem
60(12):3092–3097
190. Pandey M, Chandra DR (2015) Evaluation of ethanol and aqueous extracts of Cinnamomum
verum leaf galls for potential antioxidant and analgesic activity. Indian J Pharm Sci 77(2):243
191. Chua MT, Tung YT, Chang ST (2008) Antioxidant activities of ethanolic extracts from the
twigs of Cinnamomum osmophloeum. Bioresour Technol 99(6):1918–1925
192. Chakraborty U, Das H (2010) Antidiabetic and antioxidant activities of Cinnamomum tamala
leaf extracts in STZ-treated diabetic rats. Glob J Biotechnol Biochem 5(1):12–18
193. Anis Z, Hashim R, Hasan Mehdi S et al (2012) Radical scavenging activity, total phenol con-
tent and antifungal activity of Cinnamomum iners wood. Iran J Energy Environ 3(5):74–78
194. Park SJ, Yu MH, Kim JE, Lee SP, Lee IS (2012) Comparison of antioxidant and antimicrobial
activities of supercritical fluid extracts and marc extracts from Cinnamomum verum. J Life
Sci 22(3):373–379
195. Srinivasa RK, Kumar KN, BVV, R.K. (2012) Microwave assisted extraction and evaluation
of in vitro antioxidant activity of Cinnamomum aromaticum. J Med Plants Res 6(3):439–448
196. Abeysekera WPKM, Premakumara GAS, Ratnasooriya WD (2013) In vitro antioxidant prop-
erties of leaf and bark extracts of ceylon cinnamon (Cinnamomum zeylanicum Blume). Trop
Agric Res 24(2):128–138
197. Salleh WMNH, Farediah A, Khong HY (2015) Antioxidant and anticholinesterase activi-
ties of essential oils of Cinnamomum griffithii and C. macrocarpum. Nat Prod Commun
10(8):1465–1468
696 S. Surendran and R. Ramasubbu
219. Soni R (2013) Effect of ethanolic extract of Cinnamomum tamala leaves on wound healing
In Stz induced diabetes in rats. Asian J Pharm Clin Res 6:39–42
220. Narkhede N, Deo S, Inam F (2014) Comparative study of wound healing activity of various
spice and herbs in Rats. Int J Res Biosci Agric Tech 2(1):949–955
221. Deepa C, Srivastava R, Kumar Srivastava A et al (2016) Wound healing activity of hydro-
alcoholic extract of Cinnamomum nitidum Blume (Lauraceae) in wistar albino rats. Curr Trad
Med 2(2):134–145
222. Ahmadi SG, Farahpour MR, Hamishehkar H (2019) Topical application of Cinnamon verum
essential oil accelerates infected wound healing process by increasing tissue antioxidant
capacity and keratin biosynthesis. Kaohsiung J Med Sci 35(11):686–694
223. Kefayat A, Hamidi Farahani R, Rafienia M (2021) Synthesis and characterization of cellu-
lose nanofibers/chitosan/cinnamon extract wound dressing with significant antibacterial and
wound healing properties. J Iran Chem Soc 19:1–12
Medicinal Properties and Population
Studies on Sarcostigma kleinii Wight &
Arn.
1 Introduction
India harbours about 15% (3000–3500) out of 20,000 medicinal plants of the world.
About 90% of these are found growing wild in different climatic regions of the
country. Among these medicinal plants, Sarcostigma kleinii is a perennial large
liana. Lianas are woody climbing vines that rely on other plants for support. They
encompass 25% of species diversity in tropical systems. There is a growing body of
evidence pointing to an increase in abundance of lianas in forests. Lianas diversity
and abundance has been shown to increase following disturbance in comparisons
between forest edges and interiors and between secondary and old growth forests.
By producing many rooting stems, lianas are able to rapidly colonize disturbed
areas, thereby increasing their chances of survival [1, 2].
MS Swaminathan foundation studies reveal that over 500 species of flowering
plants are under threat in the district, of which 300 are endemic to Western Ghats
and over 50 are critically endangered with many listed in Red Data Books. Among
them, Sarcostigma kleinii is a Rare, Endemic and Threatened (RET) species and
their study reveals that Sarcostigma kleinii is a rare Iiana. Once an important source
for oil for treating rheumatism, ulcers, leprosy and skin diseases and lighting tradi-
tional lamp. (http://59.160.153.188/library/sites/default/files/25saving%2010%20
keystone%20plant%20species.pdf).
2 Family Icacinaceae
The Icacinaceae are a family of flowering plants, consisting of trees, shrubs and
lianas, primarily of the tropics. The family was traditionally circumscribed quite
broadly, with around 55 genera totaling over 400 species. In 2001, however, this
circumscription was found to be polyphyletic, and the family was split into four
families in three different orders: Icacinaceae sensu stricto, Pennantiaceae (Apiales),
Stemonuraceae (Aquifoliales) and Cardiopteridaceae (also Aquifoliales).
Icacinaceae sensu stricto contains about 150 species, distributed into about 35 gen-
era [3]. Icacina senegalensis extracts have shown activity against malaria parasites
[4]. Nothapodytes nimmoniana shows antimicrobial activity and is traditionally
used as a remedy for malaria and has anti-inflammatory activities [5]. Rhaphiostylis
beninensis root possess anti-inflammatory activities.
Ramesha et al. [6] evaluated the production of an anticancerous alkaloid camp-
tothecin (CPT) and its derivatives, in 13 species of the family Icacinaceae, namely,
Apodytes dimidiata, Codiocarpus andamanicus, Gomphandra comosa, Gomphandra
coriacea, Gomphandra polymorpha, Gomphandra tetrandra, Iodes cirrhosa, Iodes
hookeriana, Miquelia dentata, Miquelia kleinii, Natsiatum herpeticum,
Pyrenacantha volubilis and Sarcostigma kleinii. His studies were the first reports of
CPT and its derivatives in these species and offer rich alternative plant sources for
the anticancer compound, CPT. The study is also supported by [7].
Also, Shweta et al. [8] isolated the same anticancer alkaloid CPT from Miquelia
dentata Bedd. (Icacinaceae). CPT, a quinoline alkaloid, is a potent inhibitor of
eukaryotic topoisomerase I, and together with its derivatives, CPT is the third larg-
est anticancer drug in the world market. This is the first report of production of CPT
by endophytic bacteria. The identity of the bacteria was ascertained by Gram stain-
ing and 16s rRNA sequencing. This potent eukaryotic topoisomerase inhibitor,
CPT, is also reported from Pyrenacantha volubilis (Icacinaceae) from the eastern
coast of peninsular India. They analysed the CPT and its derivatives using high-
performance liquid chromatography (HPLC) coupled with electrospray mass spec-
trometry (ESI-MS) in all plant parts, such as twigs, leaves, roots, seedling, ripened
whole fruit, fruit coat, seed coat and cotyledons. Cotyledons and ripened whole
fruits contained the highest amount of CPT (1.35% and 0.60% dry weight,
respectively).
3
Sarcostigma Kleinii
Taxonomical classification
• Kingdom: Plantae
Phylum: Magnoliophyta
Class: Magnoliopsida
Order: Celastrales
Medicinal Properties and Population Studies on Sarcostigma kleinii Wight & Arn. 701
Family: Icacinaceae
Genus: Sarcostigma
Species: Sarcostigma kleinii
Vernacular names
• English: Sarcostigma kleinii
Hindi: Mukhajali
Konkani, Marathi: Davabindu
Kannada: Puvanna
Tamil: Puvennai, Ota
Malayalam: Vellayodal, Erumathali, Odal, Vattodal,
Synonyms
• Chailletia edulis Kurz
• Sarcostigma edule Kurz
• Sarcostigma horsfieldii R.Br.
• Sarcostigma roxburghii Wall. ex Griff.
• Sarcostigma wallichii Baill.
Sarcostigma kleinii belongs to the family Icacinaceae. It is a wild edible plant tradi-
tionally used by the tribes in the Parambikulam Wildlife Sanctuary, Kerala, India
[9]. The plant’s bark and leaves are bitter, acrid, thermogenic, anthelmintic, diges-
tive, carminative, diuretic, anaphrodisiac, depurative, vulnerary and stomachic.
They are useful in vitiated conditions of vata, cephalalgia, gastropathy, anorexia,
flatulence, helminthiasis, strangury, indolent ulcers, leprosy, skin diseases, hysteria
and epilepsy. The oil is bitter, anaphrodisiac, anthelmintic, vulnerary and depura-
tive. It is useful in vitiated conditions of vata, helminthiasis, foul ulcers, leprosy and
skin diseases [10] (Fig. 1).
Sarcostigma kleinii is a much-branched vigorous climbing shrub with stems up
to 26 m long and up to 8 cm thick. Usually evergreen, the plant is sometimes leafless
when flowering. The plant is used as a source of food, medicine and oil. The oil is a
popular treatment for rheumatism in India. The fruit of this plant contains a thin
sweetish pulp surrounding the single seed. Leaves and young shoots of the plant can
be used in food by cutting into small pieces and cooked with salt and chilly, and
garnished with mustard seeds, curry leaves and onions in oil.
4
Sarcostigma kleinii: Geographic Distribution
The Sarcostigma kleinii is also reported in many sacred groves of Kerala. It is avail-
able in E. Asia – southwest India, Andaman Islands, Myanmar, Vietnam, Malaya,
Indonesia. This plant is available in sacred groves. Sacred groves have been con-
served as sustainable biological resources. They serve as a valuable gene pool. They
are also considered as the first major effort to recognize and conserve biodiversity.
Since ancient period kavu has been recognized as the source of medicine, edibles
702 S. Mathew and R. Rajan
and other economically important plants for the natives. The notable medicinal spe-
cies include Anamirta cocculus, Curculigo orchioides, Cyclea peltata, Gloriosa
superba, Glycosmis pentaphylla, Ocimum spp., Phyllanthus amarus, Sarcostigma
kleinii, Scoparia dulcis and Tinospora cordifolia.
The habitat includes the Western Ghats, from Konkan southwards. It is a wild
edible plant traditionally used by the tribes in the Parambikulam Wildlife Sanctuary,
Kerala. They are usually found in Western Ghats at Low Altitudes. They are distrib-
uted widely in Maharashtra: Kolhapur, Karnataka: Chikmagalur and Coorg, Kerala:
all districts, and Tamil Nadu: Coimbatore, Kanniyakumari, Nilgiri and Tirunelveli
(http://eol.org/pages/5228816/overview).
They are woody climbers, branchlets and glabrous. Leaves are oblong-lanceolate,
apex acuminate, base rounded or obtuse; lateral nerves 8 pairs, reticulate; and peti-
ole 5–12 cm long. Flowers are 3–6 together, yellow; calyx cupular, 2 mm across, 5
toothed; petals 3–5 mm long, oblong, recurved; female flowers mostly from old
wood, ovary 1 celled, densely hairy; stigma sessile, discoid. Pistillode in male flow-
ers are conical. Fruits are drupe, orange-yellow in colour, glabrous [11] .
6 Phytochemical Constituents
7 Medicinal Properties
8 Phylogenetic Studies
Fig. 2 Phylogenetic analysis of Icacinaceae members using ndhf gene sequences. (Copyright
accessed from Ramesha et al. [6])
present study. This study evaluated the phylogeny by using maximum likelihood
analysis of Icacinaceae members sensu stricto using RAxML analysis. In the figure,
values on the nodes indicate bootstrap support for the clades and the dotted lines
species are the members of Icacinaceae sensu lato as outgroup. Numbers before the
species name indicate the GenBank accession number.
9 Conclusion
References
1. Gentry AH (1991) The distribution and evolution of climbing plants. In: Putz FE, Mooney HA
(eds) The biology of vines. Cambridge University Press, Cambridge, pp 3–52
2. Ramachandran KK, Joseph GK (2001) Feeding ecology of Nilgiri langur (Trachypithecusjohnii)
in Silent Valley National Park, Kerala, India. pp 1155–1164
3. Karehed J (2001) Multiple origin of the tropical forest tree family Icacinaceae. Am J Bot
12:2259–2274
4. Sarr SO, Perrotey S, Fall I et al (2011) Icacina senegalensis (Icacinaceae), traditionally used
for the treatment of malaria, inhibits in vitro plasmodium falciparum growth without host cell
toxicity. Malar J 10:85. https://doi.org/10.1186/1475-2875-10-85
706 S. Mathew and R. Rajan
The Editors have retracted this chapter because it has been published previously [1].
All authors agree with this retraction.
1. Krishnan, A., Joseph, J., Kalyanikutty, S. (2023). The Utility of Natural Mucilage
from the Medicinal Plant “Patha” (Cyclea peltata) as an Alternative for
Solidifying Agent in Cell Growth Media. In: Arunachalam, K., Yang, X.,
Puthanpura Sasidharan, S. (eds) Natural Product Experiments in Drug Discovery.
Springer Protocols Handbooks. Humana, New York, NY. https://doi.
org/10.1007/978-1-0716-2683-2_23
1 Introduction
About 80% of the world’s population depend upon plants or plant-based products
for food as well as health care needs as plants serve as rich source of nutrition and
safe medicines [1]. The medicinal properties of the plants can be ascribed to the
presence of various bioactive phytochemicals like phenols, flavonoids, tannins,
alkaloids and saponins [2]. The bioactive compounds that contribute to the thera-
peutic potential of the plants are actually produced by plants for their defensive
mechanism by various secondary biochemical pathways and are called secondary
metabolites [3]. They are reported to have pharmacological properties like anti-
allergic, hypoglycaemic, antioxidant, antimicrobial and anticancer potentials [4–6].
Though they can exhibit individual therapeutic effect, secondary metabolites can
work synergistically with each other and enhance pharmacological action [1]. Thus,
the medicinal value of herbs or herbal products depends on the presence and quan-
tity of phytoconstituents in them which needs to be validated by scientific methods.
Ophiorrhiza L. is a species-rich genus of the family Rubiaceae found in the wet
tropical forests of South-East Asia, extending to Australia, New Guinea and Pacific
Islands [7]. The members are generally small, herbaceous or shrubby with several
healing properties. They have been used in traditional medication as snake bite anti-
tussive and analgesic and also for curing ulcers, leprosy, gastropathy and amenor-
rhoea [8]. Ophiorrhiza brunonis Wight &Arn. is one of the several species of the
genus used in the folklore medicine for the treatment of various ailments mentioned
above. The plant is usually grown in the evergreen forest of Kerala, Tamil Nadu and
Karnataka. It is an erect herb with branched stem which is woody at the base.
Flowers are white and pubescent externally. Capsules are glabrous and green.
Though the plant is used for curing various diseases in the conventional healing
practices, the phytocomponents responsible for its medicinal properties are not elu-
cidated completely. In this background, the present study was designed for the qual-
itative and quantitative analysis of the methanolic extract of the plant. For this,
preliminary phytochemical screening, quantification of the major bioactive phyto-
components and GC–MS analysis of the plant extract were done using standard
protocols.
2 Materials
Fresh whole plant materials of O. brunonis were collected from Palakkad district of
Kerala in India. The collected plant materials were taxonomically authenticated by
Dr. M. Sabu (Professor, Department of Botany, University of Calicut, Kerala).
2.2 Chemicals Used
(1) 5% ferric chloride: 5 g FeCl3, 100 mL distilled water; (2) 10% lead acetate: 10 g
lead acetate, 100 mL distilled water; (3) Wagner’s reagent: 2 g iodine, 6 g potassium
iodide, 100 mL distilled water; (4) Hager’s reagent: Saturated picric acid solution;
(5) 1% lead acetate: 1 g lead acetate, 100 mL distilled water; (6) 1% hydrochloric
acid: 1 mL HCl, 100 mL distilled water; (7) 10% ferric chloride: 10 g FeCl3, 100 mL
distilled water; (8) 20% sodium hydroxide: 20 g NaOH, 100 mL distilled water; (9)
7% sodium carbonate: 7 g Na2CO3, 100 mL distilled water; (10) 5% sodium nitrate:
5 g Na2NO3, 100 mL distilled water; (11) Phosphate buffer (pH 4.7): 2 M sodium
phosphate, 0.2 M citric acid; (12) Bromocresol green solution; 69.8 mg BCG, 3 mL
2 N NaOH and 5 mL distilled water, make up to 1000 mL distilled water.
2.3 Equipments Used
3 Methods
The plant materials were carefully washed and shade dried. Then chopped to small
pieces and powdered. 10 g of powder was Soxhlet extracted with 100 mL of 100%
methanol for 6 to 8 h at 40–50 °C. After cooling, the extract was filtered using
Whatman filter paper No.1 and concentrated at 60 °C. The final extract was then
stored in a dark bottle at 4 °C for further experiments.
A fraction of the extract was treated with a few drops of 5% ferric chloride solution.
Formation of deep blue or black colour precipitate indicates phenol content.
About 2 mL of the extract was treated with a few drops of 10% lead acetate solution.
The formation of a yellow coloured precipitate indicates the presence of flavonoids.
About 50 mg of the extract was dissolved in dilute hydrochloric acid and then fil-
tered. The filtrate thus obtained was used for the following tests.
Wagner’s Test
Few drops of Wagner’s reagent were added to a small amount of the filtrate.
Formation of a reddish-brown precipitate indicates the presence of alkaloids.
730 S. N. Preethamol and J. E. Thoppil
Hager’s Test
The filtrate was treated with a few drops of Hager’s reagent. Appearance of orange
or yellow coloured precipitate indicates alkaloid content.
Salkowski Test
To 2 mL of the test solution, 2 mL of chloroform was added and mixed well. Few
drops of concentrated sulphuric acid were added to form a layer. A reddish-brown
colouration at the interface of the solutions indicates the presence of terpenoids.
About 0.5 g of solvent-free extract was stirred with 10 mL of distilled water and
filtered. The filtrate was analysed for the presence of tannins.
To 5 mL of the filtrate, few drops of 1% lead acetate solution were added. Yellow
precipitate indicates the presence of tannins.
Precipitation Test
Borntrager’s Test
Legal’s Test
The total phenol content of the extract was evaluated by following the methodology
of Oueslati et al. [15]. Stock concentration of the extract is prepared by dissolving
1 mg of the dried extract in 1 mL of methanol. From the stock, 0.1 mL of the extract
was taken in triplicates and mixed with 0.5 mL of distilled water and 0.125 mL of
1 N Folin-ciocalteu reagent and incubated for 5 min. After incubation, 1.25 mL of
7% sodium carbonate was added and the volume of the reaction mixture was made
up to 3 mL using distilled water. The reaction mixture was shaken well and incu-
bated in dark for another 90 min. After incubation, the absorbance of the solution
732 S. N. Preethamol and J. E. Thoppil
was measured against a blank at 760 nm. Gallic acid was the standard used. Phenolic
content of the extract was calculated from the calibration curve of the standard, and
the results were expressed as milligram of gallic acid equivalent per gram dry
weight (mg GAE/g DW) using regression equation.
The quantity of the flavonoid in the extract was determined using the method of
Oueslati et al. [15], with slight modifications. From the stock concentration of 1 mg/
mL, 0.5 mL of the extract, taken in triplicates, was mixed with 0.5 mL of distilled
water and 0.3 mL of 5% sodium nitrate and incubated for 5 min at room tempera-
ture. After incubation, 0.3 mL of 10% AlCl3 was immediately added. Thorough
shaking was followed by the addition of 2 mL sodium hydroxide (1 M). The absor-
bance was then measured at 510 nm against a suitable blank. Quercetin was used as
the standard. The TF was calculated from the calibration curve of the standard using
regression equation and was expressed in milligram of quercetin equivalent per
gram dry weight (mg QE/g DW).
Terpenoids in the extract were estimated as per Ghorai et al. [16], with slight modi-
fications. 0.2 mL of the extract (1 mg/mL), considered in triplicates, was mixed with
1.5 mL of chloroform and 1 mL of concentrated H2SO4. A yellow-green coloured
solution appeared and the chloroform layer was seen as separate layer. After sepa-
rating the chloroform layer, 2 mL of methanol was added to the residual mix which
dissolved the yellow-green solution. The absorbance of the final solution was mea-
sured against a blank at 538 nm. Linalool was the standard used. TT was calculated
from the calibration curve of linalool, and the results were expressed in milligram
of linalool equivalent per gram dry weight (mg LE/g DW).
The total alkaloid content of the extract was quantified using the protocol suggested
by Shamsa et al. [17], with slight modifications. To 1 mL of the extract (1 mg/mL),
5 mL phosphate buffer (pH 4.7) and 5 mL bromocresol green solution (BCG) were
added. To this reaction mixture, serial addition of 1, 2, 3 and 4 mL of chloroform
was done and shaken well. The BCG layer was removed and the chloroform layer
containing the alkaloid was taken for reading absorbance against a blank at 470 nm.
Caffeine was used as the standard. The TA of the extract was calculated from the
calibration curve of the standard and was expressed in milligram of caffeine equiva-
lent per gram dry weight (mg CE/g DW). The samples were analysed in triplicates.
Secondary Metabolites in Ophiorrhiza brunonis Wight & Arn. (Rubiaceae)… 733
The chemical composition of the plant extract was further analysed by GC–MS
analysis. Thermal Desorption system TD 20, fitted with WCOT column
(60 m × 0.25 mm × 0.25 m) coated with diethylene glycol was used for the analysis.
The carrier gas was helium with a flow rate of 1.21 mL/min. The column pressure
was 77.6 KPa. Injector and detector temperatures were kept at 260 °C. 6 μL of the
sample was injected and the split ratio of the column was 10:0. The parameters for
compound separation were linear temperature program of 70–260 °C at 3 °C/min
followed by 260 °C for 6 min, total run time 43 min. The MS parameters were elec-
tron ionization (EI) voltage of 70 eV, peak width of 2 s, mass range of 40–850 m/z
and detector voltage of 1.5 V.
3.5 Identification of Compounds
3.6 Statistical Analysis
All the results were statistically analysed using the software SPSS Version 20. The
data were validated using one-way analysis of variance (ANOVA) and Duncan’s
multiple range tests and those with p < 0.05 were considered statistically significant.
The results were expressed as mean ± standard error (SE).
4 Results
The phytochemical analyses of the plant extract revealed the presence of phytocon-
stituents like phenols, flavonoids, sterols, terpenoids, tannins, alkaloids, anthraqui-
nones, coumarins and glycosides. Phlobatannin was found to be absent in the
extract.
734 S. N. Preethamol and J. E. Thoppil
The TP content of the methanolic extract of O. brunonis was calculated from the
calibration curve of gallic acid and the result was observed to be 395.55 ± 0.51 mg
GAE/g DW. Also, the TF of the plant extract was calculated from the calibration
curve of quercetin and was found to be 309.83 ± 1.69 mg QE/g DW. The TT and TA
of the plant extract were calculated from the calibration curve of linalool and caf-
feine, and the results were observed to be 367.01 ± mg LE/g DW and
126.33 ± 1.36 CE/g DW, respectively.
GC–MS analysis of the plant extract revealed the presence of 13 bioactive phyto-
components belonging to different classes of compounds. The list of these com-
pounds identified from the analysis is summarised in Table 1.
The major phytoconstituents identified in the extract were terpenoids (38.77%)
such as sesquiterpenoids like neophytadiene (11.72%) and humulane-1,6-dien-3-ol
(20.66%) and triterpenoids like squalene (6.39%). 14.91% of terpene alcohols like
phytol (10.97%) and solanesol (3.94%) were also found in the extract. Three types
of sterols namely, campesterol (2.66%), stigmasterol (5.27%) and gamma sitosterol
(9.35%) were also found. n-Hexadecanoic acid (15.77%) and tetradecanoic acid
(2.57%) were the fatty acids obtained. Alkane compounds and alkaloids were also
revealed in the analysis.
Table 1 Phytocompounds identified from the methanolic extract of Ophiorrhiza brunonis using
GC–MS analysis
Molecular
Retention time Name of the compound formula Class of the compound Peak area (%)
11.185 Dodecane C12H26 Alkane 2.07
11.496 Tetradecane C14H30 Alkane 2.71
16.382 Neophytadiene C20H38 Sesquiterpenoid 11.72
17.329 n- Hexadecanoic acid C16H32O2 Fatty acid 15.77
19.168 Phytol C20H40O Diterpene alcohol 10.97
20.112 Tetradecanoic acid C14H28O2 Fatty acid 2.57
22.536 1-Anliinoisoquinoline C15H12N2 Alkaloid 5.92
26.562 Squalene C30H50 Triterpenoid 6.39
30.662 Solanesol C45H74O Terpene alcohol 3.94
33.213 Campesterol C28H48O Sterol 2.66
33.695 Stigmasterol C29H48O Sterol 5.27
35.539 gamma-Sitosterol C29H50O Sterol 9.35
41.246 Humulane-1,6-dien-3-ol C15H26O Sesquiterpenoid 20.66
Secondary Metabolites in Ophiorrhiza brunonis Wight & Arn. (Rubiaceae)… 735
Thus, from the GC–MS analysis, six different classes of compounds like terpe-
noids (38.77%), fatty acids (18.34%), sterols (17.28%), terpene alcohols (14.91%),
alkaloids (5.92%) and alkanes (4.78%) were revealed.
5 Discussion
and tetradecanoic acid were also recognised. Two terpene alcohols, namely, solane-
sol and phytol were also identified. Solanesol exhibits antimicrobial, antitumour,
anti-inflammatory and anti-ulcer activities [28]. Phytol is a very good antinocicep-
tive and antioxidant agent, which is also involved in the production of vitamins E
and K [25]. Hence, the bioactivity of all the components could be contributing
towards the healing capacity of the plant extract.
From the results, it is clear that the plant extract has many compounds with anti-
cancer properties. The major compounds identified from the GC–MS analysis, like,
humulane-1,6-dien-3-ol, squalene and solanesol are reported to possess antitumour
potential. The cumulative effect of these compounds can enhance the medicinal
properties and are a very good lead towards the anticancer potential of the plant.
6 Conclusion
The present study on the methanolic extract of O. brunonis reveals the presence of
various highly bioactive secondary metabolites. The preliminary phytochemical
screening showed the presence of different classes of metabolites, indicating the
medicinal potential of the plant. The quantitative estimation of the major bioactive
compounds suggests that the plant has commendable quantity of secondary metabo-
lites. GC–MS analysis shows that the extract has many anticancer compounds like
humulane-1,6-dien-3-ol, squalene and solanesol. This shows that the plant can be a
good candidate for research in anticancer studies. Further, compounds with anti-
inflammatory properties, antioxidant properties and antimicrobial properties were
also found. These compounds also add to the medicinal value of the plant. So the
healing efficacy of the plant might be the synergistic contribution of these com-
pounds, which probably substantiates its use in traditional medicines. The identifi-
cation of the secondary metabolites with antitumor efficacy by various analyses is a
lead toward its anticancer potential. More in vitro and in vivo trials will help to
explore more bioactive potentials of the plant as a medicine.
Acknowledgements The authors acknowledge the Forest Department of Kerala, India, for pro-
viding the permission for the collection of the plant specimens.
Conflicts of Interest The authors declare that they have no conflict of interest.
References
1. Meena AK, Bansal P, Kumar S (2009) Plants-herbal wealth as a potential source of ayurvedic
drugs. Asian J Tradit Med 4(4):152–170
2. Bajalan I, Zand M, Goodarzi M et al (2017) Antioxidant activity and total phenolic and fla-
vonoid content of the extract and chemical composition of the essential oil of Eremostachys
laciniata collected from Zagros. Asian Pac J Trop Biomed 7(2):144–146
Secondary Metabolites in Ophiorrhiza brunonis Wight & Arn. (Rubiaceae)… 737
3. Kennedy DO, Wightman EL (2011) Herbal extracts and phytochemicals: plant secondary
metabolites and the enhancement of human brain function. Adv Nutr 2(1):32–50
4. Katalinic V, Milos M, Kulisic T et al (2006) Screening of 70 medicinal plant extracts for anti-
oxidant capacity and total phenols. Food Chem 94(4):550–557
5. Mulabagal V, Tsay HS (2004) Plant cell cultures – an alternative and efficient source for the
production of biologically important secondary metabolites. Int J Appl Sci Eng 2(1):29–48
6. Borneo R, León AE, Aguirre A et al (2009) Antioxidant capacity of medicinal plants from the
Province of Córdoba (Argentina) and their in vitro testing in a model food system. Food Chem
112(3):664–670
7. Hareesh VS, Sabu M (2018) The genus Ophiorrhiza (Rubiaceae) in Andaman and Nicobar
Islands, India with a new species. Phytotaxa 383(3):259–272
8. Rajan R, Varghese SC, Kurup R et al (2013) Search for camptothecin-yielding Ophiorrhiza
species from southern Western Ghats in India: a HPTLC-densitometry study. Ind Crop Prod
43:472–476
9. Kumar GS, Jayaveera KN, Kumar CK et al (2007) Antimicrobial effects of Indian medicinal
plants against acne-inducing bacteria. Trop J Pharm Res 6(2):717–723
10. Trease GE, Evans WC (2002) Pharmacognosy, 3rd edn. Saunders Publishers, London
11. Siddiqui AA, Ali M (1997) Practical pharmaceutical chemistry. CBS Publishers and
Distributors, New Delhi, pp 126–131
12. Edeoga HO, Okwu DE, Mbaebie BO (2005) Phytochemical constituents of some Nigerian
medicinal plants. Afr J Biotechnol 4(7):685–688
13. Sofowara A (1993) Medicinal plants and traditional medicine in Africa. Spectrum Books
Ltd, Ibadan
14. Evans WC, Evans D (2002) Trease and Evans’ Pharmacognosy, 15th edn. Elsevier Health
Sciences, New York, pp 21–24
15. Oueslati S, Ksouri R, Falleh H et al (2012) Phenolic content, antioxidant, anti-inflammatory
and anticancer activities of the edible halophyte Suaeda fruticosa Forssk. Food Chem
132(2):943–947
16. Ghorai N, Chakraborty S, Gucchait S, Saha SK, Biswas S (2012) Estimation of total terpe-
noids concentration in plant tissues using a monoterpene, linalool as standard agent. Protoc
Exch. https://doi.org/10.1038/protex.2012.05
17. Shamsa F, Monsef H, Ghamooshi R et al (2008) Spectrophotometric determination of total
alkaloids in some Iranian medicinal plants. Thai J Pharm Sci 32:17–20
18. Krishnaiah D, Devi T, Bono A et al (2009) Studies on phytochemical constituents of six
Malaysian medicinal plants. J Med Plant Res 3(2):67–72
19. Mungole AJ, Awati R, Chaturvedi A et al (2010) Preliminary phytochemical screening of
Ipomoea obscura (L): a hepatoprotective medicinal plant. Int J PharmTech Res 2(4):2307–2312
20. Oyebode OA, Erukainure OL, Ibej CU et al (2019) Phytochemical constituents, antioxidant
and antidiabetic activities of different extracts of the leaves, stem and root barks of Alstonia
boonei: an in vitro and in silico study. Bot Lett 166(4):444–456
21. Tayade AB, Dhar P, Sharma M et al (2013) Antioxidant capacities, phenolic contents, and GC/
MS analysis of Rhodiola imbricata Edgew. root extracts from Trans-Himalaya. J Food Sci
78(3):402–410
22. Sengul M, Yildiz H, Gungor N et al (2009) Total phenolic content, antioxidant and antimicro-
bial activities of some medicinal plants. Pak J Pharm Sci 22(1):102–106
23. Liu RH (2004) Potential synergy of phytochemicals in cancer prevention: mechanism of
action. J Nutr 134(12):3479–3485
24. Koche D, Shirsat R, Kawale M (2016) An overview of major classes of phytochemicals: their
types and role in disease prevention. Hislopia J 9:1–11
25. Arora S, Kumar G (2017) Gas Chromatography-Mass Spectrometry (GC-MS) determination
of bioactive constituents from the methanolic and ethyl acetate extract of Cenchrus setigerus
Vahl (Poaceae). Antiseptic 2:0–31
738 S. N. Preethamol and J. E. Thoppil
26. Jiao SG, Zhang RF, Li J et al (2018) Phytochemical and pharmacological progress on
humulane-type sesquiterpenoids. China J Chin Mater Med 43(22):4380–4390
27. Reddy LH, Couvreur P (2009) Squalene: a natural triterpene for use in disease management
and therapy. Adv Drug Deliv Rev 61(15):1412–1426
28. Yan N, Liu Y, Zhang H et al (2017) Solanesol biosynthesis in plants. Molecules 22(4):510
Marine Macroalgae as a Treasure House
of Bioactive Compounds
and Nutraceuticals
Kajal Chakraborty
1 Introduction
K. Chakraborty (*)
Bioprospecting Section of Marine Biotechnology Division, Central Marine Fisheries
Research Institute, Cochin, Kerala, India
e-mail: [email protected]
Macroalgae are divided into three major classes based on different colors: green, red
and brown algae. Besides the colour other substantial differences are in their cell
wall composition, storage compounds, ultrastructure of mitosis, etc. [11]. Green
and red macroalgae were shaped by primary endosymbiosis, whereas brown algae
originated through secondary endosymbiosis [12]. The green algae belonging to the
class of Ulvophyceae are diverse with more than 900 marine originated species. A
common growth form of two widespread genera (Cladophora and Chaetomorpha)
are found as branched (or not) thin filaments. Sheets like form by two layers of cells
are typical characteristic of Ulva spp. of Ulva genus are popularly called sea lettuce
because of their appearance. A unique type of body organization of green algae is
known as siphonalean organization (or coenocytic organization). The best-known
example of siphonalean marine algae is represented by the genus Caulerpa [13]. The
phylum rhodophyta are the largest, and are classified under seven classes and thirt-
three orders [14]. Several species of red algae have branched plantlike, small bushes
like shape. There are near about 1780 species of brown algae, currently classified as
Fucophyceae (or Phaeophyceae, which includes 17 orders) of the phylum
Ochrophyta. Brown algae are widespread in all seas of the world, but the abundance
and diversity is found cold waters. The order Laminariales (of brown algae) are
specified as one of the largest (in size) among the macroalgae and are labelled with
the word kelps.
As there is an increased trend towards utilizing natural drugs rather than synthetic
drugs, the knowledge about history of traditional medicine holds an important part
to develop drugs. Consistent with the World Health Organization (WHO), around
65% of the populace has been using herbal medicines for their primary health care
[15]. However, when compared to terrestrial plants use of marine macroalgae is not
much widespread. But there are several reports over Asian countries on the utility of
algae mainly as ingredients in the cuisine. Prior to 2000 BC itself Chinese medical
literature quoted the utilization of algae for food and medicine, whereas in Western
countries red algal products, such as carrageenan, agar were extensively used as
ingredients in food products. Marine macroalgae are commonly used by humans,
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 741
and have a great economic significance. The primary use of marine macroalgae as
food and medicine was reported as early as ~14,000 years ago. These were utilized
as a binder substance for fixation of lime [16]. Despite these, they have found uses
in conventional medicine for a number of ailments (Table 1). In Central American
countries, Gracilaria species of red seaweeds have reported to possess aphrodisiac
properties [17]. Powdered Gracilaria or agar were sold as appetite modulator and
digestive health promoter in some countries.
The marine macroalgae contain a large assemblage of species that predominate in the coastal
shelf areas of Indian subcontinent
It is of note that marine life has advanced over a long period of time in austere
marine environmental situations. Remarkably, marine organisms display greater
occurrence of pharmaceutical potential, 1% vs. 0.1% (in preclinical screening) as
compared to those exhibited by the terrestrial counterparts [25], and there are
increasing reports of novel analogues of highly bioactive compounds over the
years [26].
Occurrences of marine natural products over the period from 1971 to 2015 (2)
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 743
Asian cuisine [33]. In India, Ulva, Gracilaria and Acanthophora are used in prepar-
ing food items with the coastal states of Kerala and Tamil Nadu [34–36]. Marine
macroalgae are deliberated as food supplement attributable to availability of valu-
able macro-nutrients and micro-nutrients and bioactive compounds. Nowadays,
marine macroalgae are considered as one of the major coastal resources that are
valuable to human consumption and environment in Asian, America and European
countries as ingredients in prepared foods. They are recognized for their richness in
various nutrients, such as minerals, dietary fibers and vitamins (cyanocobalamin)
other than polyunsaturated fatty acids and polyphenols [37]. The capability of
marine macroalgae to increase the storage stability of C20–22n-3 fatty acids was eval-
uated by analyzing the combined effect of organic fractions of Jania rubens, Hypnea
musciformis and Kappaphycus alvarezii [38, 39]. The potential use of seaweed spe-
cies such as Sargassum wightii, S. myriocystum, S. plagiophyllum, Caulerpa spp.,
Anthophycus longifolius, Spatoglossum asperum, Stoechospermum polypodioides,
Gracilaria corticata and Grateloupia indica as functional food supplements, have
been investigated. Sulphated polygalactans isolated from Gracilaria opuntia and
Kappaphycus alvarezii were evaluated for their potential to develop nutraceuticals.
The prebiotic activity of polysaccharide extracted from marine macroalgae
Sargassum wightii, Enteromorpha compressa, Acanthophora spicifera, etc. was
evaluated, for formulating functional food ingredients.
The number of publications in the areas of antioxidant compounds topped the list
followed by that of anti-inflammatory. The total number of publications in bioactive
compounds from marine macroalgae was deduced through google literature search
and online tools/database, such as MarinLit, Google Scholar, Science Direct,
Scifinder and PubMed. An increased number of publications in marine macroalgal
natural products acknowledged the importance of this class of marine flora.
6.1 Terpenoids
Terpenes are usually isolated from plants. Among diverse classes of drugs currently
utilized for infectious diseases and cancer treatments, around 23,000 molecules are
of terpenoid origin. Commercially available anti-malarial drug artemisnin and
746 K. Chakraborty
anti-cancer drug paclitaxel (taxol) are two fine examples in this regard. Marine mac-
roalgae are deliberated to be rich sources of terpenes, which have been isolated and
verified to possess several pharmacological potentials. According to the previous
reports, more than 1058 terpene molecules have been isolated and structurally char-
acterized only from marine macroalgae. Sargadiol-I, a terpenoid analogue, which
was isolated from Sargassum tortile, exhibited cytotoxic activity [54]. An anti-
bacterial brominated chamigrane terpene was isolated from Laurencia majus-
cula [55].
Halogenated compounds with terpenoid moieties along with other small molecu-
lar bioactives were isolated and characterized from marine macroalgae [56–61] and
were classified into terpenoids, phenols/aromatics, indoles and non-terpenoid C15-
acetogenins. Halogenated terpenoids, non-terpenoid acetogenins and indoles were
principally reported from red seaweed Laurencia, whereas the halogenated phenols
were isolated from Polysiphonia, Rhodomela and Symphyocladia [62]. Among the
halogenated compounds, halogenated terpenes and bromophenols displayed the
greater potential for new drug development [63].
6.2 Lipidic Compounds
During the previous decade, fatty acid profile of marine macroalgae has invited
much consideration by reason of their high lipid extract content of polyunsaturated
fatty acids (PUFAs) and steroids especially (18:3n-3), octadecatetranoic acid
(18:4n-3), arachidonic (20:4n-6) and eicosapentenoic acids (20:5n-3). Since marine
macroalgae are the sole organisms to synthesize long-chain PUFAs because of the
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 747
presence of specific enzymes, which are not in human, consumption of marine mac-
roalgae is highly favored. PUFAs are essential for brain development, many physi-
ological functions, signaling and regulation of transcription factors and in the
treatment of some autoimmune and cancer diseases. Red marine macroalgae were
found to contain more than 60 sterols and PUFAs and could be a sustainable source
of nutritive food. Besides, publications are not too short for their promising biologi-
cal activities, such as cytotoxicity, enzyme inhibitors, anti-inflammatory and anti-
microbial. As seaweed contains a large number of fatty acids, it provides a potential
source of raw PUFAs [9] due to the reason that these compounds restore the perme-
ability barrier and prevent skin dehydration and dermatitis [64, 65]. Deficiency of
these fatty acids could lead to alopecia and eczema [66].
Br
OH H
O
OH
Br
HO
Sargadiol-I Chamigrane
6.3 Polysaccharides
Polysaccharides are the major components of marine macroalgae, and its content
ranges between 4% and 76% of their dry weight. The marine macroalgae with high-
est polysaccharide contents include Ascophyllum, Porphyra and Palmaria [67]. Red
algae contain unique sulfated galactans, such as carrageenans agarose and agar [68,
69]. The brown algae contain sulfated polysaccharides such as laminarin, alginate
and fucoidan [70–72]. The sulphated polysaccharides exhibit many valuable bio-
logical properties like anticoagulant, antiviral, antioxidant, anti-tumour, immuno-
modulating, anti-hyperlipidaemic and anti-hepatotoxic [73].
O
O
OCH3 H
H H
O HO
6.4 Polyketide Derivatives
HO H
H O
O
Br C O H
O H H
Br
O
R1 H
H H
Cl
Polyketide derivative
[Isolaurenidificin, R1 = OH; bromlaurenidificin R1 = Br] 12-Epoxyobtusallene-IV
6.5 Polyphenolic Compounds
R
R R
R R R R
R R
O
O O
R R
R R R
R
R
R = OAc
Polyphenolic compound from brown seaweed
Various classes of bioactive compounds isolated from Padina tetrastomatica [48, 91, 92]
752 K. Chakraborty
Various classes of bioactive compounds isolated from Sargassum wightii [69, 72, 93–95]
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 753
7.1 Miscellaneous
Table 2 (continued)
Pharmacological
Terpenes/Terpenoids Algal species property References
Five sesquiterpenes Dictyopteris Anti-cancer [125]
Oplopane sesquiterpene divaricata
Perhydroazulene diterpene Dictyota Algicidal activity [126]
dichotoma
Two diterpenes Dictyota sp. Anti-tuberculosis [127]
Anti-malarial
Cytotoxic
Dolabellane diterpene Dictyota pfaffii Anti-fouling [128]
Seven meroditerpenoids and three Cystoseira Anti-fouling [129]
derivatives baccata Anti-bacterial
Four meroditerpenoids Halidrys Anti-fouling [130]
A chromene siliquosa
Seven dolabellanes Dilophus spiralis Anti-bacterial [131]
Amijiol acetate Dictyota Cytotoxic [132]
Dolabellatrienol dichotoma Protective against
Amijiol-7,10-diacetate DNA damage
Three meroterpenoids Sargassum Anti-oxidant [133]
siliquastrum
One dolabellane Dictyota pfaffii Anti-viral [116]
Sargachromanol Q Sargassum Cytotoxic [134]
Sargachromanol R siliquastrum
Seco-Taondiol Stypopodium Gastroprotective [135]
flabelliforme activity
Cystophloroketals A−E Cystoseira Anti-fouling [136]
tamariscifolia
Cadinan-4(15)-ene-1β,5α-diol Dictyopteris – [137]
Trans-3-norisocalamenen-4-ol divaricata
Bifurcatriol Bifurcaria Anti-protozoal activity [138]
bifurcata
Two dolastane diterpenes Canistrocarpus Cytotoxic [139]
cervicornis
Two xenicin diterpenoids Padina Anti-inflammatory [47]
Three xeniolide-type diterpenoids tetrastomatica
Three dolabellanes Padina Anti-diabetic [47]
Two dolastanes tetrastromatica
Sargilicixenicane Sargassum Anti-oxidant [95]
ilicifolium Anti-inflammatory
Two hydroperoxysterols Turbinaria ornata Cytotoxic [140]
3β,28ξ-dihydroxy-24-ethylcholesta- Sargassum Cytotoxic [141]
5,23Z-dien carpophyllum
2a-oxa-2- oxo-5α-hydroxy-3,4-dinor-
24-ethylcholesta-24(28)-ene
Sulfoquinovosyldiacylglycerol Lobophora Anti-protozoal [142]
variegata
(continued)
756 K. Chakraborty
Table 2 (continued)
Pharmacological
Terpenes/Terpenoids Algal species property References
Cymatherelactone Cymathere Sodium channel [143]
cymatherols A-C methyl ester triplicata blocking
derivatives
Spiralisones A–D Zonaria spiralis Anti-bacterial [144]
Neurodegenrative
kinase inhibition
Dictyoptesterols A–C Dictyopteris PTP1Binhibitory [145]
undulata activity
Dictyopterisins A–J Dictyopteris PTP1Binhibitory [146]
undulata activity
Cytotoxic
Two polyketide lactones Sargassum Anti-oxidant [147]
wightii Anti-inflammatory
Eckstolonol Ecklonia Anti-oxidant [148]
stolonifera
Phloroglucinol derivative Eisenia bicyclis Anti-diabetic [149]
Phlorofucofuroeckol Eisenia arborea Anti-allergic [150]
Fucodiphlorethol G Ecklonia cava Anti-oxidant [151]
Diphlorethohydroxycarmalol Ishige okamurae Anti-diabetic [152]
Two phloroglucinols Fucus vesiculosus Anti-oxidant [153]
cytochromeP450
enzyme inhibition
Eckmaxol Ecklonia maxima Neuroprotective [154]
effects
Deoxylapachol Landsburgza Anti-fungal [155]
querciflia Cytotoxic
Diphlorethohydroxycarmalol Ishige okamurae Cytotoxic [156]
5′-Deoxy-5′-methylamino-adenosine Laminaria Growth stimulant [157]
japonica
Pheophytin a Sargassum Neurodiffrentiation [158]
fulvellum compound
Apo-9′-fucoxanthinone Cladostephus – [159]
Apo-13′-fucoxanthinoneloliolide spongiosus
Comosusols A–D Sporochnus Cytotoxic [160]
Comosone A comosus
produced in 2016. Carrageenan and agar from red seaweed are used as additives for
various products, such as water based gelling desserts, ice cream, cheeses, low-
calorie jellies, soy milk, flan puddings, dairy products, chocolate milk and processed
and canned meat, jellies, seasonings, beer sauces and other processed foods.
Gelidium, Gracilaria, Gelidiella and Pterocladiella are the species mostly used for
the extraction of gelatin which is a solidifying agent and an additive in confection
of pie fillings and toppings.
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 757
Marine macroalgae contain a large assemblage of species that predominate in the coastal
shelf areas of Indian subcontinent
Over the past few years, use of marine macroalgae to develop value-added com-
pounds and novel pharmacophores has attracted attention from pharmaceutical and
food industries. Nutraceutical products have been developed from marine macroal-
gae as green alternatives to synthetic drugs to combat rheumatic arthritic pains and
type-2 diabetes. Antihypercholesterolemic and antihypothyroidism nutraceuticals
were also developed from marine macroalgae to combat dyslipidaemia and hypo-
thyroid disorder. Lead molecules with action against angiotensin-converting
enzyme-I from marine macroalgae were isolated and added to a nutraceutical prod-
uct [40–42, 147, 162, 163].
9 Conclusions
Marine macroalgae are attaining tremendous attention due to the presence of bioac-
tive polysaccharides, terpenes, alkaloids, steroids, fatty acids, polyphenols and pig-
ments. These compounds are representatives of several biological properties such as
anti-oxidant, anti-inflammatory and anti-diabetic . From the nutritional point of
view, they are very rich in minerals, vitamins and soluble dietary fiber contents. Till
now, there are many species that remains unexplored and undoubtedly there opens
a wild field of research into the discovery and development of new natural product
based nutraceuticals and drugs. The diverse natural compounds from marine mac-
roalgae with unique medicinal activities has introduced algal products into pharma-
ceutical industry. Recent efforts to produce clearness in regulatory protocols in
connection with the nutraceutical products have also increased growth of this seg-
ment of marine microalgal research and their utilization to develop high-value com-
pounds and products for human health perspective. As a step towards upholding the
utilization of marine macroalgae, cohesive aquaculture of these species has been
commenced. The idea of utilization of marine macroalgae as sea vegetables via low-
technology approach in third world countries is very approachable and practical. In
addition to this, development of functional food products also could provide oppor-
tunities to popularize the health benefits of marine macroalgae. Apart from agar and
carrageenan, a number of pharmaceutically important constituents can be isolated
from marine macroalgae which opens an alternative strategy to fulfill the market’s
demand for naturally derived compounds in food and medicines.
758 K. Chakraborty
Acknowledgements The author thanks the Director, Central Marine Fisheries Research Institute
for facilitating the research activities. This work was funded by the Indian Council of Agricultural
Research, New Delhi, India under Central Marine Fisheries Research Institute supported project
“Development of Bioactive Pharmacophores from Marine Organisms” (grant number MBT/
HLT/SUB23).
References
1. Kong DX, Jiang YY, Zhang HY (2010) Marine natural products as sources of novel scaffolds:
achievement and concern. Drug Discov Today 15(21–22):884–886. https://doi.org/10.1016/j.
drudis.2010.09.002
2. Blunt JW, Copp BR, Keyzers RA, Munro MH, Prinsep MR (2016) Marine natural products.
Nat Prod Rep 33(3):382–431. https://doi.org/10.1039/c5np00156k
3. Blunt JW, Carroll AR, Copp BR, Davis RA, Keyzers RA, Prinsep MR (2018) Marine natural
products. Nat Prod Rep 35(1):8–53. https://doi.org/10.1039/c7np00052a
4. Xie J, Zhang AH, Sun H, Yan GL, Wang XJ (2018) RSC Adv 8:812–824
5. Šimat V, Elabed N, Kulawik P, Ceylan Z, Jamroz E, Yazgan H, Čagalj M, Regenstein JM,
Özogul F (2020) Recent advances in marine-based nutraceuticals and their health benefits.
Mar Drugs 18(12):627. https://doi.org/10.3390/md18120627
6. Carroll AR, Copp BR, Davis RA, Keyzers RA, Prinsep MR (2021) Marine natural products.
Nat Prod Rep 38:362–413. https://doi.org/10.1039/D0NP00089B
7. Driggers EM, Hale SP, Lee J, Terrett NK (2008) The exploration of macrocycles for drug
discovery- an underexploited structural class. Nat Rev Drug Discov 7(7):608–624. https://
doi.org/10.1038/nrd2590
8. Faulkner DJ (2001) Marine natural products. Nat Prod Rep 18(1):1–49. https://doi.
org/10.1039/b006897g
9. Khotimchenko SV, Vaskovsky VE, Titlyanova TV (2002) Fatty acids of marine algae from the
Pacific coast of North California. Bot Mar 45:17–22. https://doi.org/10.1515/BOT.2002.003
10. Kishida R, Yamagishi K, Muraki I, Sata M, Tamakoshi A, Iso H, JACC study group (2020)
Frequency of seaweed intake and its association with cardiovascular disease mortality: the
jacc study. J Atheroscler Thromb 27(12):1340–1347. https://doi.org/10.5551/jat.53447
11. Rindi F, Pasella MM, Lee ME, Verbruggen H (2020) Phylogeography of the mediterranean
green seaweed Halimeda tuna (Ulvophyceae, Chlorophyta). J Phycol 56(4):1109–1113.
https://doi.org/10.1111/jpy.13006
12. Rindi F, Soler Vila A, Guiry M (2012) Taxonomy of marine macroalgae used as sources
of bioactive compounds. Mar Bioact Compd. https://doi.org/10.1007/978-1-4614-1247-2_1
13. Stam WT, Olsen JL, Zaleski SF, Murray SN, Brown KR, Walters LJ (2006) A forensic and
phylogenetic survey of Caulerpa species (Caulerpales, Chlorophyta) from the Florida coast,
local aquarium shops, and e-commerce: establishing a proactive baseline for early detection.
J Phycol 42(5):1113–1124. https://doi.org/10.1111/j.1529-8817.2006.00271.x
14. Saunders GW, Hommersand MH (2004) Assessing red algal supraordinal diversity and tax-
onomy in the context of contemporary systematic data. Am J Bot 91(10):1494–1507. https://
doi.org/10.3732/ajb.91.10.1494
15. Fabricant DS, Farnsworth NR (2001) The value of plants used in traditional medicine for drug
discovery. Environ Health Perspect 109(1):69–75. https://doi.org/10.1289/ehp.01109s169
16. Chengkui Z, Tseng CK, Junfu Z, Chang CF (2004) Chinese seaweeds in herbal medicine.
Hydrobiologia 116-117:152–154. https://doi.org/10.1007/BF00027655
17. Featonby-Smith BC, Van Stade J (1983) The effect of seaweed concentrate on the growth of
tomato plants in nematode infested soil. Sci Hortic 20:137–147
18. Nadkarni AK, Nadkarni KM (1996) Nadkarni’s Indian materia medica: with Ayurvedic,
Unani- Tibbi, Siddha, allopathic, homeopathic, naturopathic & home remedies, appendices
& indexes, 3rd edn. Popular Prakashan, Mumbai
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 759
19. Fonnegra R, Jiménez SL (2007) Gracilaria (alga rodofícea), Plantas medicinales aprobadas
en Colombia, 2nd edn. Editoral Universidad de Antioquía, Medellín, pp 124–125
20. Anggadiredja JT (2009) Ethnobotany study of seaweed diversity and its utilization in
Warambadi, Panguhalodo areas of East Sumba district. Jurnal Teknologi Lingkungan:297–310.
https://doi.org/10.29122/jtl.v10i3.1476
21. Chengkui Z (1984) Phycological research in the development of the Chinese seaweed indus-
try. Hydrobiologia 116:7–18. https://doi.org/10.1007/BF00027633
22. Watt G (2014) A dictionary of the economic products of India, Gossypium to Linociera, vol
4. Cambridge University Press. https://doi.org/10.1017/CBO9781107239173
23. FAO (2020) The state of world fisheries and aquaculture. Rome, 244 p
24. Andrade PB, Barbosa M, Matos RP, Lopes G, Vinholes J, Mouga T, Valentão P (2013)
Valuable compounds in macroalgae extracts. Food Chem 138(2–3):1819–1828. https://doi.
org/10.1016/j.foodchem.2012.11.081
25. Munro MH, Blunt JW, Dumdei EJ, Hickford SJ, Lill RE, Li S, Battershill CN, Duckworth AR
(1999) The discovery and development of marine compounds with pharmaceutical potential.
J Biotechnol 70(1–3):15–25. https://doi.org/10.1016/s0168-1656(99)00052-8
26. Montaser R, Luesch H (2011) Marine natural products: a new wave of drugs? Future Med
Chem 3(12):1475–1489. https://doi.org/10.4155/fmc.11.118
27. Chakraborty K, Antony T (2021) First report of spiro-compounds from marine macroalga
Gracilaria salicornia: prospective natural anti-inflammatory agents attenuate 5-lipoxygenase
and cyclooxygenase-2. Nat Prod Res 35(5):770–781. https://doi.org/10.1080/1478641
9.2019.1608545
28. McHugh DJ (2003) A guide to the seaweed industry. FAO Fisheries Technical Paper 441.
Food and Agriculture Organization of the United Nations, Rome
29. Ganesan AR, Tiwari U, Rajauria G (2019) Seaweed nutraceuticals and their therapeutic role
in disease prevention. Food Sci Human Wellness 8(3):252–263
30. Tsuji S (1980) Japanese cooking: a simple art. Kodansha America, Incorporated,
New York, 518 p
31. Fujii M (2005) The enlightened kitchen. Kodansha International, Tokyo, 107 p
32. Griffin J (2015) An investigative study into the beneficial use of seaweed in bread and the
broader food industry. Dissertation presented to Dublin Institute of Technology, School of
Culinary Arts and Food Technology in partial fulfillment of the requirements for the bach-
elor’s degree BSc (Hons) Baking and Pastry Arts Management, 132 p
33. Bocanegra A, Bastida S, Benedí J, Ródenas S, Sánchez-Muniz FJ (2009) Characteristics and
nutritional and cardiovascular-health properties of seaweeds. J Med Food 12(2):236–258.
https://doi.org/10.1089/jmf.2008.0151
34. Dhargakar VK (2014) Uses of seaweeds in the Indian diet for sustenance and well-being. Sci
Cult 80(7–8):192–202
35. Subba RPV, Ganesan K, Suresh KK (2009) Seaweeds: a survey of research and utiliza-
tion. In: Khattar JIS, Singh DP, Gurpreet Kaur IK (eds) Algal biology and biotechnology.
International Publishing House, Pvt. Ltd, New Delhi/Bangalore, pp 165–178
36. Subba RPV, Periyasamy C, Rama RK, Srinivasa RA (2016) Seaweed for human welfare.
Seaweed Res Util 38:1–12
37. FAO (2003) The state of world fisheries and aquaculture. Rome
38. Chakraborty K, Joseph D (2018) Effects of antioxidative substances from seaweed on quality
of refined liver oil of leafscale gulper shark, Centrophorus squamosus during an accelerated
stability study. Food Res Int 103:450–461. https://doi.org/10.1016/j.foodres.2017.10.018
39. Chakraborty K, Joseph D (2018) Effect of antioxidant compounds from seaweeds on storage
stability of C20-22 polyunsaturated fatty acid concentrate prepared from dogfish liver oil. Food
Chem 260:135–144. https://doi.org/10.1016/j.foodchem.2018.03.144
40. Chakraborty K et al (2010) A process to prepare antioxidant and anti-inflammatory con-
centrates from brown and red seaweeds and a product thereof. Indian Patent Grant number
294451 (2064/CHE/2010, granted on 16/03/2018)
41. Chakraborty K et al (2012) A product containing anti-inflammatory principles from brown
seaweeds and a process thereof. Indian Patent number 5199/CHE/2012
760 K. Chakraborty
42. Chakraborty K. (2015) A process to prepare antidiabetic concentrates from seaweeds and a
product thereof. Indian Patent Appl. no. 3366/DEL/2015
43. Chakraborty K. (2017) A process to prepare anti-dyslipidemic concentrate from seaweed and
a product thereof. Indian Patent Appl. no. 201711013741
44. Chakraborty K, Joseph D (2016) Antioxidant potential and phenolic compounds of brown
seaweeds Turbinaria conoides and Turbinaria ornata (class: Phaeophyceae). J Aquat Food
Prod Technol 25(8):1249–1265. https://doi.org/10.1080/10498850.2015.1054540
45. Chakraborty K et al (2020) A composition and antihypertensive product from marine algae.
Indian Patent Appl no. 202011011489
46. Makkar F, Chakraborty K (2018) Highly oxygenated antioxidative 2H-chromen deriva-
tive from the red seaweed Gracilaria opuntia with pro-inflammatory cyclooxygenase and
lipoxygenase inhibitory properties. Nat Prod Res 32(23):2756–2765. https://doi.org/10.108
0/14786419.2017.1378209
47. Antony T, Chakraborty K (2019) Xenicanes attenuate pro-inflammatory 5-lipoxygenase:
prospective natural anti-inflammatory leads from intertidal brown seaweed Padina tetrastro-
matica. Med Chem Res 28:591–607. https://doi.org/10.1007/s00044-019-02322-8
48. Antony T, Chakraborty K (2020) First report of antioxidative 2H-chromenyl derivatives from
the intertidal red seaweed Gracilaria salicornia as potential anti-inflammatory agents. Nat
Prod Res 34(24):3470–3482. https://doi.org/10.1080/14786419.2019.1579807
49. Antony T, Chakraborty K, Joy M (2021) Antioxidative dolabellanes and dolastanes from
brown seaweed Padina tetrastromatica as dual inhibitors of starch digestive enzymes. Nat
Prod Res 35(4):614–626. https://doi.org/10.1080/14786419.2019.1591402
50. Pati MP, Sharma SD, Lakshman N, Panda CR (2016) Uses of seaweed and its applica-
tion to human welfare: a review. Int J Pharm Pharm Sci 8:12–20. https://doi.org/10.22159/
ijpps.2016v8i1012740
51. Bilal M, Iqbal HMN (2019) Marine seaweed polysaccharides-based engineered cues for the
modern biomedical sector. Mar Drugs 18(1):7. https://doi.org/10.3390/md18010007
52. Blunt JW, Copp BR, Keyzers RA, Munro MH, Prinsep MR (2014) Marine natural products.
Nat Prod Rep 31(2):160–258. https://doi.org/10.1039/c3np70117d
53. Newman DJ, Cragg GM (2012) Natural products as sources of new drugs over the 30 years
from 1981 to 2010. J Nat Prod 75(3):311–335. https://doi.org/10.1021/np200906s
54. Numata A, Kanbara S, Takahashi C, Fujiki R, Yoneda M, Usami Y, Fujita E (1992) A cytotoxic
principle of the brown alga Sargassum tortile and structures of chromenes. Phytochemistry
31(4):1209–1213. https://doi.org/10.1016/0031-9422(92)80262-D
55. Vairappan CS, Daitoh M, Suzuki M, Abe T, Masuda M (2001) Antibacterial halogenated
metabolites from the Malaysian Laurencia species. Phytochemistry 58(2):291–297. https://
doi.org/10.1016/s0031-9422(01)00243-6
56. Rocha DHA, Seca AML, Pinto DCGA (2018) Seaweed secondary metabolites: in vitro and
in vivo anticancer activity. Mar Drugs 16(11):410. https://doi.org/10.3390/md16110410
57. Makkar F, Chakraborty K (2018) Antioxidant and anti-inflammatory oxygenated meroterpe-
noids from the thalli of red seaweed Kappaphycus alvarezii. Med Chem Res 27(8):2016–2026.
https://doi.org/10.1007/s00044-018-2210-0
58. Makkar F, Chakraborty K (2018) Antioxidative sulphated polygalactans from marine mac-
roalgae as angiotensin-I converting enzyme inhibitors. Nat Prod Res 32(17):2100–2106.
https://doi.org/10.1080/14786419.2017.1363756
59. Makkar F, Chakraborty K (2018) First report of dual cyclooxygenase-2 and 5-lipoxygenase
inhibitory halogen derivatives from the thallus of intertidal seaweed Kappaphycus alvarezii.
Med Chem Res 27:2331–2340. https://doi.org/10.1007/s00044-018-2239-0
60. Makkar F, Chakraborty K (2018) Novel furanyl derivatives from the red seaweed Gracilaria
opuntia with pharmacological activities using different in vitro models. Med Chem Res
27:1245–1259. https://doi.org/10.1007/s00044-018-2144-6
61. Makkar F, Chakraborty K (2018) Previously undescribed antioxidative azocinyl morpho-
linone alkaloid from red seaweed Gracilaria opuntia with anti-cyclooxygenase and lipox-
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 761
96. Hori K, Matsubara K, Miyazawa K (2000) Primary structures of two hemagglutinins from
the marine red alga, Hypnea japonica. Biochim Biophys Acta 1474(2):226–236. https://doi.
org/10.1016/s0304-4165(00)00008-8
97. Gauto DF, Di Lella S, Estrin DA, Monaco HL, Martí MA (2011) Structural basis for ligand
recognition in a mushroom lectin: solvent structure as specificity predictor. Carbohydr Res
346(7):939–948. https://doi.org/10.1016/j.carres.2011.02.016
98. Manning JC, Romero A, Habermann FA, García Caballero G, Kaltner H, Gabius HJ (2017)
Lectins: a primer for histochemists and cell biologists. Histochem Cell Biol 147(2):199–222.
https://doi.org/10.1007/s00418-016-1524-6
99. Cardoso SM, Pereira OR, Seca AM, Pinto DC, Silva AM (2015) Seaweeds as preven-
tive agents for cardiovascular diseases: from nutrients to functional foods. Mar Drugs
13(11):6838–6865. https://doi.org/10.3390/md13116838
100. Kim SK, Pangestuti R (2011) Biological activities and potential health benefits of fucoxanthin
derived from marine brown algae. Adv Food Nutr Res 64:111–128. https://doi.org/10.1016/
B978-0-12-387669-0.00009-0
101. Bedoux G, Hardouin K, Burlot AS, Bourgougnon N (2014) Bioactive components from sea-
weeds: cosmetic applications and future development. Adv Bot Res 71:345–378. https://doi.
org/10.1016/B978-0-12-408062-1.00012-3
102. Pereira L (2018) Seaweeds as source of bioactive substances and skin care therapy – cos-
meceuticals, algotheraphy, and thalassotherapy. Cosmetics 5:68. https://doi.org/10.3390/
cosmetics5040068
103. Salehi B, Sharifi-Rad J, Seca AML, Pinto DCGA, Michalak I, Trincone A, Mishra AP,
Nigam M, Zam W, Martins N (2019) Current trends on seaweeds: looking at chemical com-
position, phytopharmacology, and cosmetic applications. Molecules 24(22):4182. https://doi.
org/10.3390/molecules24224182
104. Fadli M, Aracil JM, Jeanty G, Banaigs B, Francisco C (1991) Novel meroterpenoids from
Cystoseira mediterranea: use of the crown-gall bioassay as a primary screen for lipophilic
antineoplastic agents. J Nat Prod 54(1):261–264. https://doi.org/10.1021/np50073a029
105. Rovirosa J, Sepulveda M, Quezada E, San-Martin A (1992) Isoepitaondiol, a diterpenoid of
Stypopodium flabelliforme and the insecticidal activity of stypotriol, epitaondiol and deriva-
tives. Phytochemistry 31(8):2679–2681. https://doi.org/10.1016/0031-9422(92)83610-B
106. Kurata K, Taniguchi K, Suzuki M (1996) Cyclozonarone, a sesquiterpene-substituted
benzoquinone derivative from the brown alga Dictyopteris undulata. Phytochemistry
41(3):749–752. https://doi.org/10.1016/0031-9422(95)00651-6
107. Davyt D, Enz W, Manta E, Navarro G, Norte M (1997) New chromenols from
the brown alga Desmarestia menziesii. Nat Prod Rep 9(4):305–312. https://doi.
org/10.1080/10575639708043644
108. Wessels M, König GM, Wright AD (1999) A new tyrosine kinase inhibitor from the marine
brown alga Stypopodium zonale. J Nat Prod 62(6):927–930. https://doi.org/10.1021/
np990010h
109. Bennamara A, Abourriche A, Berrada M, Charrouf M, Chaib N, Boudouma M, Garneau
FX (1999) Methoxybifurcarenone: an antifungal and antibacterial meroditerpenoid from the
brown alga Cystoseira tamariscifolia. Phytochemistry 52(1):37–40. https://doi.org/10.1016/
s0031-9422(99)00040-0
110. Ayyad SE, Slama MO, MoKhtar AH, Anter AF (2001) Cytotoxic bicyclic diterpene from the
brown alga Sargassum crispum. Boll Chim Farm 140(3):155–159
111. Takada N, Watanabe R, Suenaga K, Yamada K, Uemura D (2001) Isolation and structures
of hedaols A, B, and C, new bisnorditerpenes from a Japanese brown alga. J Nat Prod
64(5):653–655. https://doi.org/10.1021/np0005661
112. Fisch KM, Böhm V, Wright AD, König GM (2003) Antioxidative meroterpenoids from the
brown alga Cystoseira crinita. J Nat Prod 66(7):968–975. https://doi.org/10.1021/np030082f
113. Gedara SR, Abdel-Halim OB, El-Sharkawy SH, Salama OM, Shier TW, Halim AF (2003)
Cytotoxic hydroazulene diterpenes from the brown alga Dictyota dichotoma. Z Naturforsch
C: Biosci 58(1–2):17–22. https://doi.org/10.1515/znc-2003-1-203
764 K. Chakraborty
114. Soto H, Rovirosa J, San-Martin A (2003) A new diterpene from Dictyota crenulata. Z
Naturforsch B: J Chem Sci 58(8):795–798. https://doi.org/10.1515/znb-2003-0812
115. Siamopoulou P, Bimplakis A, Iliopoulou D, Vagias C, Cos P, Vanden Berghe D, Roussis
V (2004) Diterpenes from the brown algae Dictyota dichotoma and Dictyota linearis.
Phytochemistry 65(14):2025–2030. https://doi.org/10.1016/j.phytochem.2004.06.018
116. Barbosa JP, Pereira RC, Abrantes JL, Cirne dos Santos CC, Rebello MA, Frugulhetti IC,
Texeira VL (2004) In vitro antiviral diterpenes from the Brazilian brown alga Dictyota pfaffii.
Planta Med 70(9):856–860. https://doi.org/10.1055/s-2004-827235
117. Pereira HS, Leão-Ferreira LR, Moussatché N, Teixeira VL, Cavalcanti DN, Costa LJ, Diaz R,
Frugulhetti IC (2004) Antiviral activity of diterpenes isolated from the Brazilian marine alga
Dictyota menstrualis against human immunodeficiency virus type 1 (HIV-1). Antivir Res
64(1):69–76. https://doi.org/10.1016/j.antiviral.2004.06.006
118. Song FH, Fan X, Xu XL, Zhao JL, Han LJ, Shi JG (2004) A new sesquiterpene-substituted
benzoic acid from the brown alga Dictyopteris divaricata. Chin Chem Lett 15(3):316–318.
https://doi.org/10.1080/1028602032000169532
119. Jang KH, Lee BH, Choi BW, Lee HS, Shin J (2005) Chromenes from the brown alga
Sargassum siliquastrum. J Nat Prod 68(5):716–723
120. Mori J, Iwashima M, Wakasugi H, Saito H, Matsunaga T, Ogasawara M, Takahashi S, Suzuki
H, Hayashi T (2005) New plastoquinones isolated from the brown alga, Sargassum micra-
canthum. Chem Pharm Bull (Tokyo) 53(9):1159–1163. https://doi.org/10.1248/cpb.53.1159
121. Abatis D, Vagias C, Galanakis D, Norris JN, Moreau D, Roussakis C, Roussis V (2005)
Atomarianones A and B: two cytotoxic meroditerpenes from the brown alga Taonia atom-
aria. Tetrahedron Lett 46(49):8525–8529. https://doi.org/10.1016/j.tetlet.2005.10.007
122. Tziveleka LA, Abatis D, Paulus K, Bauer R, Vagias C, Roussis V (2005) Marine polyprenyl-
ated hydroquinones, quinones, and chromenols with inhibitory effects on leukotriene forma-
tion. Chem Biodivers 2(7):901–909. https://doi.org/10.1002/cbdv.200590066
123. Sabry OM, Andrews S, McPhail KL, Goeger DE, Yokochi A, LePage KT, Murray TF,
Gerwick WH (2005) Neurotoxic meroditerpenoids from the tropical marine brown alga
Stypopodium flabelliforme. J Nat Prod 68(7):1022–1030. https://doi.org/10.1021/np050051f
124. Song F, Xu X, Li S, Wang S, Zhao J, Cao P, Yang Y, Fan X, Shi J, He L, Lü Y (2005)
Norsesquiterpenes from the brown alga Dictyopteris divaricata. J Nat Prod 68(9):1309–1313.
https://doi.org/10.1021/np040227y
125. Song F, Fan X, Xu X, Zhao J, Yang Y, Shi J (2006) Cadinane sesquiterpenes from the
brown alga Dictyopteris divaricata. J Nat Prod 67(10):1644–1649. https://doi.org/10.1021/
np040099d
126. Kim JY, Alamsjah MA, Hamada A, Fujita Y, Ishibashi F (2006) Algicidal diterpenes from the
brown alga Dictyota dichotoma. Biosci Biotechnol Biochem 70(10):2571–2574. https://doi.
org/10.1271/bbb.60281
127. Jongaramruong J, Kongkam N (2007) Novel diterpenes with cytotoxic, anti-malarial and anti-
tuberculosis activities from a brown alga Dictyota sp. J Asian Nat Prod Res 9(6–8):743–751.
https://doi.org/10.1080/10286020701189203
128. Barbosa J, Fleury B, Da Gama B, Teixeira V, Pereira R (2007) Natural products as antifou-
lants in the Brazilian brown alga Dictyota pfaffii (Phaeophyta, Dictyotales). Biochem Syst
Ecol 35:549–553. https://doi.org/10.1016/j.bse.2007.01.010
129. Mokrini R, Mesaoud MB, Daoudi M, Hellio C, Maréchal JP, El Hattab M, Ortalo-Magné
A, Piovetti L, Culioli G (2008) Meroditerpenoids and derivatives from the brown alga
Cystoseira baccata and their antifouling properties. J Nat Prod 71(11):1806–1811. https://
doi.org/10.1021/np8004216
130. Culioli G, Ortalo-Magné A, Valls R, Hellio C, Clare AS, Piovetti L (2008) Antifouling
activity of meroditerpenoids from the marine brown alga Halidrys siliquosa. J Nat Prod
71(7):1121–1126. https://doi.org/10.1021/np070110k
131. Ioannou E, Quesada A, Rahman MM, Gibbons S, Vagias C, Roussis V (2011) Dolabellanes
with antibacterial activity from the brown alga Dilophus spiralis. J Nat Prod 74(2):213–222.
https://doi.org/10.1021/np1006586
Marine Macroalgae as a Treasure House of Bioactive Compounds and Nutraceuticals 765
132. Ayyad SE, Makki MS, Al-Kayal NS, Basaif SA, El-Foty KO, Asiri AM, Alarif WM, Badria
FA (2011) Cytotoxic and protective DNA damage of three new diterpenoids from the brown
alga Dictoyota dichotoma. Eur J Med Chem 46(1):175–182. https://doi.org/10.1016/j.
ejmech.2010.10.033
133. Lee SH, Kang SM, Ko SC, Lee DH, Jeon YJ (2012) Octaphlorethol A, a novel phenolic com-
pound isolated from a brown alga, Ishige foliacea, increases glucose transporter 4-mediated
glucose uptake in skeletal muscle cells. Biochem Biophys Res Commun 420(3):576–581.
https://doi.org/10.1016/j.bbrc.2012.03.036
134. Lee JI, Park BJ, Kim H, Seo Y (2014) Isolation of two new meroterpenoids from
Sargassum siliquastrum. Bull Korean Chem Soc 35:2867–2869. https://doi.org/10.5012/
bkcs.2014.35.9.2867
135. Areche C, Benites J, Cornejo A, Ruiz LM, García-Beltrán O, Simirgiotis MJ, Sepúlveda
B (2015) Seco-taondiol, an unusual meroterpenoid from the Chilean seaweed Stypopodium
flabelliforme and its gastroprotective effect in mouse model. Mar Drugs 13(4):1726–1738.
https://doi.org/10.3390/md13041726
136. El Hattab M, Genta-Jouve G, Bouzidi N, Ortalo-Magné A, Hellio C, Maréchal JP, Piovetti L,
Thomas OP, Culioli G (2015) Cystophloroketals A-E, unusual phloroglucinol-meroterpenoid
hybrids from the brown alga Cystoseira tamariscifolia. J Nat Prod 78(7):1663–1670. https://
doi.org/10.1021/acs.jnatprod.5b00264
137. Ji NY, Song YP, Miao FP, Liang XR (2016) Three cadinane derivatives from the marine
brown alga Dictyopteris divaricata. Magn Reson Chem 54(1):88–90. https://doi.org/10.1002/
mrc.4319
138. Smyrniotopoulos V, Merten C, Kaiser M, Tasdemir D (2017) Bifurcatriol, a new antipro-
tozoal acyclic diterpene from the brown alga Bifurcaria bifurcata. Mar Drugs 15(8):245.
https://doi.org/10.3390/md15080245
139. Campbell S, Murray J, Delgoda R, Gallimore W (2017) Two new oxodolastane diterpenes
from the Jamaican macroalga Canistrocarpus cervicornis. Mar Drugs 15(6):150. https://doi.
org/10.3390/md15060150
140. Shen YC, Tsai PI, Fenical W, Hay ME (1992) Secondary metabolite chemistry of the
caribbean marine alga Sporochnus bolleanus: a basis for herbivore chemical defence.
Phytochemistry 32(1):71–75
141. Tang HF, Yang-Hua Y, Yao XS, Xu QZ, Zhang SY, Lin HW (2002) Bioactive steroids from
the brown alga Sargassum carpophyllum. J Asian Nat Prod Res 4(2):95–101. https://doi.
org/10.1080/10286020290027362
142. Cantillo-Ciau Z, Moo-Puc R, Quijano L, Freile-Pelegrín Y (2010) The tropical brown alga
Lobophora variegata: a source of antiprotozoal compounds. Mar Drugs 8(4):1292–1304.
https://doi.org/10.3390/md8041292
143. Choi H, Proteau PJ, Byrum T, Pereira AR, Gerwick WH (2012) Cymatherelactone and
cymatherols A-C, polycyclic oxylipins from the marine brown alga Cymathere triplicata.
Phytochemistry 73(1):134–141. https://doi.org/10.1016/j.phytochem.2011.09.014
144. Zhang H, Xiao X, Conte MM, Khalil Z, Capon RJ (2012) Spiralisones A-D: acylphloroglu-
cinol hemiketals from an Australian marine brown alga, Zonaria spiralis. Org Biomol Chem
10(48):9671–9676. https://doi.org/10.1039/c2ob26988k
145. Yang F, Zhang LW, Feng MT, Liu AH, Li J, Zhao TS, Lai XP, Wang B, Guo YW, Mao SC
(2018) Dictyoptesterols A-C, ∆22-24-oxo cholestane-type sterols with potent PTP1B inhibi-
tory activity from the brown alga Dictyopteris undulata Holmes. Fitoterapia 130:241–246.
https://doi.org/10.1016/j.fitote.2018.09.001
146. Feng MT, Wang T, Liu AH, Li J, Yao LG, Wang B, Guo YW, Mao SC (2018) PTP1B inhibi-
tory and cytotoxic C-24 epimers of Δ28-24-hydroxy stigmastane-type steroids from the brown
alga Dictyopteris undulata Holmes. Phytochemistry 146:25–35
147. Maneesh A, Chakraborty K (2017) Unprecedented antioxidative and anti-inflammatory aryl
polyketides from the brown seaweed Sargassum wightii. Food Res Int 100(1):640–649.
https://doi.org/10.1016/j.foodres.2017.07.006
766 K. Chakraborty
148. Kang HS, Chung HY, Jung JH, Son BW, Choi JS (2003) A new phlorotannin from the
brown alga Ecklonia stolonifera. Chem Pharm Bull (Tokyo) 51(8):1012–1014. https://doi.
org/10.1248/cpb.51.1012
149. Okada Y, Ishimaru A, Suzuki R, Okuyama T (2004) A new phloroglucinol derivative from the
brown alga Eisenia bicyclis: potential for the effective treatment of diabetic complications. J
Nat Prod 67(1):103–105. https://doi.org/10.1021/np030323j
150. Sugiura Y, Matsuda K, Yamada Y, Nishikawa M, Shioya K, Katsuzaki H, Imai K, Amano H
(2006) Isolation of a new anti-allergic phlorotannin, phlorofucofuroeckol-B, from an edible
brown alga, Eisenia arborea. Biosci Biotechnol Biochem 70(11):2807–2811. https://doi.
org/10.1271/bbb.60417
151. Ham YM, Baik JS, Hyun JW, Lee NH (2007) Isolation of a new phlorotannin, fucodiphlor-
ethol G, from a brown alga Ecklonia cava. Bull Korean Chem Soc 28(9):1595
152. Heo SJ, Hwang JY, Choi JI, Han JS, Kim HJ, Jeon YJ (2009) Diphlorethohydroxycarmalol
isolated from Ishige okamurae, a brown algae, a potent alpha-glucosidase and alpha-
amylase inhibitor, alleviates postprandial hyperglycemia in diabetic mice. Eur J Pharmacol
615(1–3):252–256. https://doi.org/10.1016/j.ejphar.2009.05.017
153. Parys S, Kehraus S, Krick A, Glombitza KW, Carmeli S, Klimo K, Gerhäuser C, König GM
(2010) In vitro chemopreventive potential of fucophlorethols from the brown alga Fucus
vesiculosus L. by anti-oxidant activity and inhibition of selected cytochrome P450 enzymes.
Phytochemistry 71(2–3):221–229. https://doi.org/10.1016/j.phytochem.2009.10.020
154. Wang J, Zheng J, Huang C, Zhao J, Lin J, Zhou X, Naman CB, Wang N, Gerwick W, Wang
Q, Cui W, He S (2018) Eckmaxol, a Phlorotannin extracted from Ecklonia maxima, produces
anti-β-amyloid oligomer neuroprotective effects possibly via directly acting on glycogen
synthase kinase 3β. ACS Chem Neurosci 9. https://doi.org/10.1021/acschemneuro.7b00527
155. Perry NB, Blunt JW, Munro MH (1991) A cytotoxic and antifungal 1,4-naphthoquinone and
related compounds from a New Zealand brown algae, Landsburgia quercifolia. J Nat Prod
54(4):978–985. https://doi.org/10.1021/np50076a009
156. Toume K, Miyata M, Egawa K, Nose K, Hayashi M, Komiya K, Ishibashi M (2004) Isolation
of diphlorethohydroxycarmalol from a brown alga Ishige okamurae. J Nat Med 58(2):79–80
157. Kuwada K, Kuramoto M, Utamura M, Matsushita I, Ishii T (2006) Isolation and structural
elucidation of a growth stimulant for arbuscular mycorrhizal fungus from Laminaria japon-
ica Areschoug. J Appl Phycol 18(6):795–800. https://doi.org/10.1007/s10811-006-9095-x
158. Ina A, Hayashi K, Nozaki H, Kamei Y (2007) Pheophytin a, a low molecular weight com-
pound found in the marine brown alga Sargassum fulvellum, promotes the differentiation of
PC12 cells. Int J Dev Neurosci 25(1):63–68. https://doi.org/10.1016/j.ijdevneu.2006.09.323
159. El Hattab M, Culioli G, Valls R, Richou M, Piovetti L (2008) Apo-fucoxanthinoids and loliolide
from the brown alga Cladostephus spongiosus f. verticillatus (Heterokonta, Sphacelariales).
Biochem Syst Ecol 36(5–6):447–451. https://doi.org/10.1016/j.bse.2007.08.016
160. Ovenden SP, Nielson JL, Liptrot CH, Willis RH, Wright AD, Motti CA, Tapiolas DM (2011)
Comosusols A-D and comosone A: cytotoxic compounds from the brown alga Sporochnus
comosus. J Nat Prod 74(4):739–743. https://doi.org/10.1021/np1008009
161. Subudhi S (2017) Bioprospecting for algal based nutraceuticals and high value added com-
pounds. J Pharm Pharm. https://doi.org/10.15436/2377-1313.17.1651
162. Chakraborty K et al (2019) An oral drug delivery system and an anti-bacterial topical appli-
cant from seaweed based hydrogel and a process thereof. Indian patent Application No
201911018958
163. Chakraborty K. et al (2020) A composition and anti-osteoporotic product from seaweeds.
Indian patent Application No 202011009121
Plant Metabolites as New Leads to Herbal
Drug Discovery: Approaches
and Challenges
1 Introduction
Traditional medicines and natural goods are quite important. For many years, natu-
ral compounds and derivatives have been recognised as a source of medicinal agents
and structural variety. Natural products have a wide range of multidimensional
chemical structures, and their efficacy as biological function modifiers has also
received a lot of attention.
Finding robust and viable lead candidates, which is nothing more than the pro-
cess flow from a screening of natural product to a novel isolate, is proving to be a
difficult scientific work that demands skill and experience. However, in addition to
their chemical structure diversity and biodiversity, modern technologies have trans-
formed natural product screening in the search for novel medications. The use of
these technologies presents a once-in-a-lifetime opportunity to reinstate natural
products as a significant source of drug discovery.
Kapish Kapoor, Priyal Jain, and Joohee Pradhan died before publication of this work was
completed.
The primary stage of chemical isolation entails identifying the specific plant species
that provide medical benefits. The essential components in that plant species that
are linked to druggability are screened out and separated in the secondary stage.
Chemistry, biology, biochemistry, pharmacology, and history are just a few of the
subjects covered. Andrographolide from Andrographispaniculata, Picroside from
Picrorrhizakurroa, Morphine from Papaversomniferum, and Berberine from
Berberisaristata are examples of successful ethnomedicinal approaches.
Several chemicals, such as L-Dopa and paclitaxel, have been effectively extracted
from plants, such as Mucunaprurita and Taxusbrevifolia, regardless of their ethno-
medical utility.
Strategies for drug development from natural source:
The conventional approach makes advantage of the vast array of botanical sources
available as remedies. Being a common medical strategy, it differs from the ethno-
medicinal system in the following ways:
(a) It is generally acknowledged and used, as opposed to the ethnomedicinal sys-
tem, which is used by a small number of families within a community.
(b) Unlike ethnomedicinal approach, the pharmacological procedure is fully
defined and standardised.
(c) While ethnomedicine is a simply cumulative examination of therapy, the tradi-
tional approach includes an analytical relationship between experimental con-
clusions and human physiological character and pharmacological principles [13].
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 769
Table 1 Herbal drugs with active constituent, source, and synthetic analogues
Metabolite Botanical
S. No. Disease class Molecule source Synthetic analogues
1 Anti-cancer Alkaloid Catharanthus Catharanthus Navelbine (vinorelbine),
[1–3] roseus Oncovin (vincristine),
Velban (vinblastine)
Alkaloid Taxol Taxus brevifolia Docetaxel
Alkaloid Camptothecin Camptotheca Topotecan and
acuminata Irinotecan
Lignan Podophyllotoxin Podophyllum Teniposide
pelatum,
Podophyllum
emodi
2 Anti- Alkaloid Belladona Atropa Glycopyrrolate
cholinergic belladonna,
[4, 5] Datura metel,
Alkaloid Physotigmine Physostigma Phenserine
venenosum
Alkaloid Pilocarpine Pilocarpus N-methyl derivative
jaborandi containing (S)-3-ethyl
4-[(4,-imidazolyl)
methyl]-2 oxazolidinone
3 Anti- Alkaloid Reserpine Rauvolfia Reserpine methonitrate
hypertensive serpentine
[6]
4 Anti-malarial Alkaloid Cinchona Cinchona sp. Artemisinin
[4]
5 Anti-gout Alkaloid Colchicine Colchicum Desacetylmethyl
[7] autumnale colchicin
(colcimid) and
Demecolcin
6 Local Alkaloid Cocaine Erythroxylum R-pseudococaine,
Anaesthetic coca Salicylmethylecgonine,
[4, 8] Methylvanillylecgonine
7 Skeletal Alkaloid d-turbocurarine Strychnos –
muscle toxifera,
relaxant Chondodendron
[9]
8 Analgesics Alkaloid Opium Papaver –
[10] somniferum
9 Contraceptives Steroid Diosgenin Dioscorea spp. Diosgenone
[11]
10 Cardiotonic Steroid Digoxin Digitalis –
[4, 12] purpurea
770 K. Kapoor et al.
2.1.2 Ethno-Pharmacology Approach
Plants with various health advantages have been identified as a result of close inves-
tigation of cow grazing patterns and behavioural patterns. Cattles attracted to a spe-
cific plant species were a sign that the plant possessed certain properties that were
good to the health and well-being of its users. In some parts of Southern America,
such cattle grazing habits led to the discovery of plant species belonging to the
Solanaceae family, which are high in vitamin D3 derivatives [13].
2.2.1 Parallel Approach
When the biological activity of a plant is well recognised, either through traditional
use or indigenous knowledge, this strategy is used. The major goal of this screening
procedure is to extract the specific component from the plant that is responsible for the
plant‘s biological activity or medical significance. The strategy is laid out in Fig. 1 [13].
Plant Extraction
Biological Fractionation
evaluation
Druggabilty
Test
Plant
Extract Hexane Chloroform Ethyl Acetate Butanol Aqueous
BIOLOGICAL ASSAY
COMPOUNDS
STRUCTURAL CHARACTERISATION
DRUGGABILITY TEST
2.2.2 Step-by-Step Approach
This method is an alternative to the parallel screening method. It’s frequently used
in ad hoc tactics or when the plant‘s specific biological activity is unknown. The
method is depicted in Fig. 2 as a flow diagram [13].
O
O
2 Isoelemicin
O
O
O
3 3-(4-Allyl-2,6-Dimethoxyphenoxy)-2,3-Dihydro-5-Methoxy-2-Methylbenzofuran
O
O O
O
4 Myristicin
O
5 Surinamensin
O
O
O
O O
OH
6 Malabaricone
OH O
OCH3
OH
OH
7 5-Methoxylicarin A
OCH3
OH
O
O O
8 Methoxylicarin A
O
O
O
O
O O O
(continued)
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 773
Table 2 (continued)
Sr. No Compound
9 Licarin A
H
OH
O
O O
10 Malabaricone B
OH O
H
OH
OH
11 Licarine C
OCH3
OCH3
O
O O
12 5-Methoxylicarin B
OCH3
O
O
O
O
13 Licarin B
H
O
O
O
O
3.2 Gas Chromatography
3.3 Spectroscopic Techniques
3.3.1 Ultraviolet-Visible Spectroscopy
Uv-vis regions range from wavelengths 190 nm to 800 nm, and almost all organic
molecules are transparent under this region. Bioactive compounds can be analysed
through UV VIS spectrophotometer in isolated or in mixture form. Some of the
phytochemicals detected are total phenolic extract (280 nm), flavones (320 nm),
phenolic acids (360 nm), etc. [19].
Organic molecules contain certain functional groups known as chromophore
having valence electrons. These electrons of each functional group absorbs UV-vis
radiation at different wavelengths and thus a certain molecule have a number of
absorption band in absorption spectrum.These aborption bands corresponds to
structural groups within the molecule. 95%ethanol is the most suitable solvent used
because most classes of compounds solubilises in it. other solvents used are water,
ether, petroleum, hexane, and methanol [17].
3.3.2 Infrared Spectroscopy
Infrared spectroscopy used IR radiation are the most useful region that ranges
between 4000 and 670 cm−1. Only those molecules can absorb irradiation whose
vibrational changes within the molecule causes net change in the dipole moment.
Many functional groups can be identified by their characteristic vibration frequen-
cies. It has a role in structural elucidation where new compounds are identified in
plants [17]. The absorptions of each type of bond (NIH, CIH, OIH, CIX, CJO, CIO,
CIC, CJC, CKC, CKN, and so on) are regularly found only in certain small portions
of the vibrational infrared region. A small range of absorption can be defined for
each type of bond. Outside this range, absorptions are normally due to some other
type of bond [20]. Therefore, the structure of a bioactive compound can be deter-
mined by detecting the characteristic frequency absorption band for the functional
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 775
HO O HO O
OH
OH OH
OH O OH O
QUERCETIN MYRICETIN
O O H
H H
HO
2 Sitosterol H
6 Siderol
H H
H H H H
HO HO
3 Stigmasterol 10 Xanthomicrol
OH
O
H OAc
HO CH2OH
4 Campesterol 11 Penduletin
OH OH
OCH3 H
H3CO H3CO
H3CO H3CO
H OCH3
OH O
OH O
NMR served as a powerful and a rapid tool both to navigate the targeted isolation
process of bioactive constituents and reveal the identity of bioactive components.1D
1
H NMR spectroscopy was applied in the crude extract to monitor the presence of
siderol and identify other secondary metabolites. Henceforth, a total of 11 com-
pounds were revealed from the chemical investigation of dichloromethane extract of
S. euboea aerial parts, including one fatty acid ester, 2-(p-hydroxyphenyl) ethyl
stearate; three phytosterols, namely sitosterol, stigmasterol, and campesterol; one
triterpenoid, ursolic acid; four diterpenoids, i.e. siderol, eubol, eubotriol,
7-epicandicandiol; and two flavonoids, xanthomicrol and penduletin as depicted in
Table 4. Another example of NMR utilisation reported were the 1H NMR spectra of
Agave syrups and comparative sweeteners were obtained. 1H NMR data coupled to
multivariate methods such as principal component analysis (PCA) allowed the iden-
tification and classification of Agave syrups as well as differentiation with respect to
other sweeteners [24].
3.3.4 Mass Spectroscopy
The pharmaceutical industry uses mass spectrometry in all phases of the drug devel-
opment process, from lead compound discovery and structural analysis, to synthetic
development and combinatorial chemistry, and to pharmacokinetics and drug
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 777
HO O
OH
OH O
2 Caffeic acid
O
OH
HO
OH
3 Myricetin
OH
OH
HO O
OH
OH
OH O
778 K. Kapoor et al.
As previously said, global flora has endowed humanity with several medicinal com-
pounds. What has been uncovered thus far is just the tip of the iceberg. Many more
have yet to be discovered; however, as explained in the following sections, several
obstacles stand in the way of more innovative medications being discovered from
plant sources.
and ease of use in liquid systems. The test material must not be degraded, coagu-
lated, precipitated, or inert to undesirable chemical reactions in order to execute
HTS correctly. Plant metabolites or plant extracts, on the other hand, frequently fail
to meet any, if not all, of these characteristics. Plant samples, for example, are
extremely viscous; they tend to coagulate and, if left undisturbed, precipitate out of
the solution. As a result, maintaining homogeneity in the HTS system is difficult.
Another point to consider is that HTS observations and outcomes are frequently
fluorescence dependant. In general, plant samples now contain fluorescent particles
or fluorescence quenching chemicals, which contribute to HTS colorimetric end-
points being misled [48]. Apart from that, plant extracts contain a variety of highly
polarised and highly non-polarised chemicals, such as polyphenols and flavonoids,
which can interfere with the HTS test type reaction. Even chlorophyll falls into this
category [49]. In addition to organic components, inorganic substances such as
heavy metal salts, which plants have a tendency to acquire from the environment,
can induce false-positive or false-negative results in HTS [50].
5.2 Demand Factor
Plant-based raw materials for the creation of new medicinal compounds are in high
demand right now. The global demand for medicinal plants is increasing at a high
rate of almost 15–20% per year, with the Indian market valued at around $1 billion
each year [51].
However, the increased demand for medicinal plants has resulted in the over-
harvesting of several plant species that contain medicinal plant components.
Taxusbaccata, for example, has been gathered in large quantities from Himalayan
wild forests in order to extract the taxol molecule, which is particularly successful in
the treatment of ovarian cancer. Plants such as Gloriosa superba (flame lily), Aconitum
heterophyllum, Arnebia benthamii, Dactylorhiza hatagirea, Megacarpoea polyandra,
and others have shown similar behaviour. They have been over-harvested and over-
used due to high demand and are now classified as endangered species. Certain
medicinal plants that are powerful against various diseases have been over-collected
and exploited to the point where they are on the verge of extinction in the wild [52].
Aegle marmelos is a plant that can be used to treat 31 ailments. Hemidescus indicus,
on the other hand, is utilised to treat 34 different disorders. Because of the excessive
use of the target plants, they were quickly cut down, reducing their availability and
supply in the wild. Simultaneously, the genetic diversity of those locations was dis-
rupted, affecting the natural ecosystem and livelihoods of the residents [53].
5.3 Increasing Rarity
The continued overuse of beneficial plant species with strong therapeutic potential
resulted in population declines of those specific target species in their native areas.
This resulted in a significant loss of habitat and livelihood for those who rely on
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 781
them. The erosion of shared social practises for conserving and preserving natural
resources is the primary cause of danger. Weakening of customary laws due to vari-
ous powerful socio-economic forces, as well as several other threatening factors
such as habitat alteration, climatic changes, invasion of non-natives, overgrazing of
livestock, high-density population, genetic drift, interruption in land usage, patho-
gens, and predators, have limited the high availability and posed a threat to the
abundance of rare medicinal plant species in that geographical area [52].
In some circumstances, different species of the same genera can be used to treat
the same ailment; for example, different Swertia sp., such as Swertia augustifolia,
Swertia cordata, Swertia chirajyta, and others, can be used to cure malarial fever.
As a result, there is no undue stress on a particular Swertia sp. On the other side,
species like Rauvolfia serpentine are under a lot of stress because they do not have
any other load-sharing partners. As a result, the risk of extinction and hazard associ-
ated with increasing rarity is a serious concern for those species that have metabo-
lites that are not typically found in other species, as in the case of R. serpentine [54].
5.4 Threat of Extinction
Rather than taking portions of an interesting plant from the forest, it would have
been preferable if those plants could be farmed. This will eliminate the threat of
extinction, as well as the biodiversity and forest preservation issues. However, there
is extremely little information about plant propagation (only 10%), and agrotech-
nology has only been created successfully for 1% of the plants [57]. As a result, it is
reasonable to believe that most medicinal plants cannot be adequately cultivated due
to a lack of knowledge. As a result, agro-technology development must be one of
the most important research fields. This invention would aid in meeting rising mar-
ket demand while maintaining product quality consistency [54].
782 K. Kapoor et al.
Agro-forestry may be another option, as it has been observed that the forest envi-
ronment aids in the growth of many medicinal plants and the production of certain
metabolites with therapeutic properties. The use of agroforestry can benefit us in the
following ways:
(a) In a multi-strata system, shade-tolerant medicinal plants can be used in the
lower strata, giving us the possibility to produce.
(b) As a secondary crop, short-cycle therapeutic plants can be grown.
(c) In agro-forestry, larger trees with medicinal advantages can give shade and
serve as boundary markers.
(d) Medicinal plants and food crops can be interplanted [53].
Although the investment in medicinal plant production is more than that of typical
food crops, the return is also lower. This is the primary reason why most farmers are
uninterested in growing medicinal plants. Several organisations are attempting to
cultivate several therapeutic plants that are uncommon or endangered. They
attempted to produce 20 endangered medicinal plant species from northern India,
but only 10 were successful, with Rheum emobi being the most economically effi-
cient [58, 59].
5.6 Bio-piracy
regulate and monitor but also rendering them ineffective. Plant metabolites have
gained in relevance as a result of better understanding of these issues [63]. Another
key strategy is now being implemented. Researchers are choosing for semisynthetic
or mimicking natural metabolites rather than entirely natural or synthetic molecules
[64]. Scientists have identified favoured structures in some special compounds with
specialised structures that are relatively common plant metabolites, such as benzo-
diazepines [65], N-acylhydrazone [66], and indoles [67].
These privileged structures are now the subject of inquiry. They are creating new
compounds based on semisynthetic or entirely synthetic architectures. To some
extent, mimicking these specific structures also yields favourable benefits. Newman
[68, 69] explains the various ways for these approaches in detail.
7 Conclusion
There are immense opportunities to investigate the therapeutic and other biological
aspects of previously inaccessible natural items as there is a growing interest in
herbal medication development with minimal adverse effects. It is necessary to con-
centrate on visualisation and identification of underused herbal plants all over the
world in order to determine its utility. The emphasis is therefore placed on the ratio-
nal and scientific extraction, separation, and characterisation of phytochemicals,
which are a gift from nature. The exploitation of natural products for the benefit of
humanity and the development of novel leads for drug discovery are both unmet
needs. After obtaining the phytochemical, it can be used for further research via
QSAR studies, molecular modelling, and animal studies, followed by a clinical
study. The potential of natural products to treat various ailment, as promising drug
candidates remain an important approach towards novel drug discovery to the
researcher and pharmaceutical companies. Natural goods have complicated chemi-
cal structures that vary depending on the species, and when existing high-tech pro-
cedures are used, new pharmaceuticals can be discovered, benefiting the entire
planet. As a result, the globe is always provided with nature, and man is gifted with
brain, so let us use it to discover new entities that will be available to the general
public at a reasonable cost, and we will be glad to live on this planet. Natural prod-
ucts have also been and will continue to be valuable sources of novel pharmacologi-
cal molecules. According to references found in literature, natural plants were used
to make life possible or prolong it many years ago. No life on Earth is feasible in the
twenty-first century without herbal pharmaceuticals or items obtained via natural
herbal drug development.
We examined the ways researchers use to obtain plant metabolites with drug-
gability at the opening of this chapter. These methods are simple to use, but when
we consider the amount of plants that still need to be tested for medicinal purposes,
they can become a daunting process.
As a result, several protocols can be investigated. The main issue we have men-
tioned with plant metabolites is that they are frequently incompatible with modern
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 785
References
1. Lee ON, Ak G, Zengin G, Cziáky Z, Jekő J, Rengasamy KRR et al (2020) Phytochemical com-
position, antioxidant capacity, and enzyme inhibitory activity in callus, Somaclonal variant,
and Normal green shoot tissues of Catharanthus roseus (L) G. Don. Molecules 25(21):4945
2. Rutterschmidt D, Kovács Z, Lorantfy L, Misek Z, Rajsch G (2015) Taxol purification with
centrifugal partition chromatography. Planta Med 81(16):PW_152
3. Wang X, Tanaka M, Krstin S, Peixoto HS, Moura CCM, Wink M (2016) Cytoskeletal inter-
ference – a new mode of action for the anticancer drugs camptothecin and topotecan. Eur J
Pharmacol 789:265–274
4. Balandrin MF, Kinghorn AD, Farnsworth NR (1993) Plant-derived natural products in drug
discovery and development. Human medicinal agents from plants, ACS symposium series,
534. American Chemical Society, pp 2–12
5. Zhan ZJ, Bian HL, Wang JW, Shan WG (2010) Synthesis of physostigmine analogues and
evaluation of their anticholinesterase activities. Bioorg Med Chem Lett 20(5):1532–1534
6. Landazuri P, Loango N, Restrepo B (2017) Medicinal plants used in the management hyper-
tension. J Anal Pharm Res 5
7. Kumar A, Singh B, Sharma PR, Bharate SB, Saxena AK, Mondhe DM (2016) A novel micro-
tubule depolymerizing colchicine analogue triggers apoptosis and autophagy in HCT-116
colon cancer cells. Cell Biochem Funct 34(2):69–81
8. Mcleod IK (2017) Local anesthetics. Medscape. Mechanism of action: hormones with intra-
cellular receptors
9. Waldman HJ (1994) Centrally acting skeletal muscle relaxants and associated drugs. J Pain
Symptom Manag 9(7):434–441
10. Fields A, Wiegand TJ (2014) Opium and the constituent opiates. In: Wexler P (ed) Encyclopedia
of toxicology, 3rd edn. Academic Press, Oxford, pp 698–701
11. Raymon LP (2013) Pharmacology and mechanism of action of drugs. In: Siegel JA, Saukko
PJ, Houck MM (eds) Encyclopedia of forensic sciences, 2nd edn. Academic Press, Waltham,
pp 210–217
786 K. Kapoor et al.
12. Woods KL (1986) The mode of action of cardiac glycosides. J Clin Hosp Pharm 11(1):11–13
13. Katiyar C, Gupta A, Kanjilal S, Katiyar S (2012) Drug discovery from plant sources: an inte-
grated approach. Ayu 33(1):10–19
14. Ganesan A (2008) The impact of natural products upon modern drug discovery. Curr Opin
Chem Biol 12(3):306–317
15. Sasidharan S, Chen Y, Dharmaraj S, Karupiah S, Lachimanan Y (2011) Extraction, isolation
and characterization of bioactive compounds from plants? Extracts. Afr J Tradit Complement
Altern Med 8:1–10
16. Chiu S, Wang T, Belski M, Abourashed EA (2016) HPLC-guided isolation, purification and
characterization of Phenylpropanoid and phenolic constituents of nutmeg kernel (Myristica
fragrans). Nat Prod Commun 11(4):1934578X1601100416
17. Shah B, Seth A (2012) Textbook of pharmacognosy and phytochemistry – E-book. Elsevier
Health Sciences
18. Konappa N, Udayashankar AC, Krishnamurthy S, Pradeep CK, Chowdappa S, Jogaiah S
(2020) GC–MS analysis of phytoconstituents from Amomum nilgiricum and molecular dock-
ing interactions of bioactive serverogenin acetate with target proteins. Sci Rep 10(1):16438
19. Sharma M, Kaushik P (2021) Vegetable phytochemicals: an update on extraction and analysis
techniques. Biocatal Agric Biotechnol 36:102149
20. Pavia D, Lampman G, Kriz G, Vyvyan J (2015) Introduction to spectroscopy. In: Pavia DL,
Lampman GM, Kriz GS, Vyvyan JR (eds) A small scale approach to organic laboratory tech-
niques. Cengage Learning
21. Rakhee MJ, Sharma RK, Misra K (2018) Chapter 9 – Characterization techniques for
herbal products. In: Misra K, Sharma P, Bhardwaj A (eds) Management of High Altitude
Pathophysiology. Academic Press, pp 171–202
22. Bashir RA (2020) Phytochemical screening and Fourier transformed infrared spectroscopy(FT-
IR) analysis of Vernonia amygdalina del. Methanol Leaf Extract
23. Hazra K (2019) Phytochemical investigation of terminalia bellirica fruit inside. J Pharm Clin
Res 12(8):191–194
24. Velázquez Ríos IO, González-García G, Mellado-Mojica E, Veloz García RA, Dzul Cauich
JG, López MG et al (2019) Phytochemical profiles and classification of agave syrups using (1)
H-NMR and chemometrics. Food Sci Nutr 7(1):3–13
25. Barupal T, Meena M, Sharma K (2019) Inhibitory effects of leaf extract of Lawsonia inermis
on Curvularia lunata and characterization of novel inhibitory compounds by GC–MS analysis.
Biotechnol Rep 23:e00335
26. Padma MGS, Jayaseelan T, Azhagumadhavan S, Sasikala P, Senthilkumar S, Mani P (2019)
Phytochemical screening and GC–MS analysis of bioactive compounds present in ethanolic
leaves extract of Silybum marianum (L). JDDT [Internet] 9(1):85–89
27. Shakya A, Correspondence A (2016) Medicinal plants: future source of new drugs. Int J Herb
Med 4:59–64
28. Bernhoft A (2010) A brief review. In: Bernhoft A (ed) Bioactive compounds in plants—ben-
efits and risks for man and animals. The Norwegian Academy of Science and Letters, Oslo,
pp 11–17
29. Nema R, Khare S, Jain P, Pradhan A, Gupta A, Singh D (2013) Natural products potential and
scope for modern cancer research. Am J Plant Sci 4:1270–1277
30. Kaur R, Kapoor K, Kaur HP (2011) Plants as a source of anticancer agents. J Nat Prod Plant
Resour 1:119–124
31. Kumar S, Mishra A, Pandey AK (2013) Antioxidant mediated protective effect of Parthenium
hysterophorus against oxidative damage using in vitro models. BMC Complement Altern Med
13(1):120
32. Khazir J, Mir BA, Pilcher L, Riley DL (2014) Role of plants in anticancer drug discovery.
Phytochem Lett 7:173–181
33. Prakash O, Kumar A, Kumar P, Ajeet A (2013) Anticancer potential of plants and natural prod-
ucts: a review. Am J Pharmacol Sci 1(6):104–115
Plant Metabolites as New Leads to Herbal Drug Discovery: Approaches and Challenges 787
34. Cragg GM, Boyd MR, Cardellina JH 2nd, Newman DJ, Snader KM, McCloud TG (1994)
Ethnobotany and drug discovery: the experience of the US National Cancer Institute. Ciba
Found Symp 185:178–190; discussion 90-6
35. Rao PV, Nallappan D, Madhavi K, Rahman S, Jun Wei L, Gan SH (2016) Phytochemicals
and biogenic metallic nanoparticles as anticancer agents. Oxidative Med Cell Longev
2016:3685671
36. Greenwell M, Rahman PKSM (2015) Medicinal plants: their use in anticancer treatment. Int J
Pharm Sci Res 6(10):4103–4112
37. Lohani A, Verma A, Joshi H, Yadav N, Karki N (2014) Nanotechnology-based cosmeceuticals.
ISRN. Dermatology 2014:843687
38. Alonso-Castro AJ, Villarreal ML, Salazar-Olivo LA, Gomez-Sanchez M, Dominguez F,
Garcia-Carranca A (2011) Mexican medicinal plants used for cancer treatment: pharmacologi-
cal, phytochemical and ethnobotanical studies. J Ethnopharmacol 133(3):945–972
39. Jena J, Gupta AK (eds) (2012) Ricinus communis linn: a phytopharmacological review
40. Pashaei-Asl F, Pashaei-Asl R, Khodadadi K, Akbarzadeh A, Ebrahimie E, Pashaiasl M (2018)
Enhancement of anticancer activity by silibinin and paclitaxel combination on the ovarian
cancer. Artif Cells Nanomed Biotechnol 46(7):1483–1487
41. Umadevi M, Rajeswari R, Rahale CS, Selvavenkadesh S, Pushpa R, Kumar KPS,
Bhowmik D (2012) Traditional and medicinal uses of Withania Somnifera. Pharm Innov
1(9):102–110
42. Gajalakshmi S, Rajeswari D (2013) Pharmacological activities of Catharanthus roseus: A per-
spective review. Int J Pharm Biosci 4:431–439
43. Sharma V, Sarkar IN (2013) Bioinformatics opportunities for identification and study of
medicinal plants. Brief Bioinform 14(2):238–250
44. Akhtar MS, Swamy MK (2018) Anticancer plants: natural products and biotechnological
implements, vol 2. Springer, Singapore
45. Rathinamoorthy R, Thilagavathi G (2014) Terminalia chebula – review on pharmacological
and biochemical studies. Int J PharmTech Res 6:97–116
46. Raina H, Soni G, Jauhari N, Sharma N, Bharadvaja N (2014) Phytochemical importance of
medicinal plants as potential sources of anticancer agents. Turk J Bot 38:1027–1035
47. Song Y-H, Sun H, Zhang A, Yan G-L, Han Y, Wang X-J (eds) (2014) Plant-derived natural
products as leads to anti-cancer drugs
48. Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, Linder T, Wawrosch C, Uhrin P et al
(2015) Discovery and resupply of pharmacologically active plant-derived natural products: a
review. Biotechnol Adv 33(8):1582–1614
49. Zhou J, Du G, Chen J (2014) Novel fermentation processes for manufacturing plant natural
products. Curr Opin Biotechnol 25:17–23
50. Fernando DR, Marshall AT, Forster PI, Hoebee SE, Siegele R (2013) Multiple metal accumu-
lation within a manganese-specific genus. Am J Bot 100(4):690–700
51. Joshi K, Chavan P, Warude D, Patwardhan B (2004) Molecular markers in herbal drug technol-
ogy. Curr Sci 87(2):159–165
52. Kala CPFN, Dhar U (2005) Traditional uses and conservation of Timur (Zanthoxylum arma-
tum DC.) through social institutions in Uttaranchal Himalaya. India Conservat Soc 3:224–230
53. Rao MR, Palada MC, Becker BN (2004) Medicinal and aromatic plants in agroforestry sys-
tems. Agrofor Syst 61(1):107–122
54. Kala CP, Dhyani PP, Sajwan BS (2006) Developing the medicinal plants sector in northern
India: challenges and opportunities. J Ethnobiol Ethnomed 2(1):32
55. Cordell GA (2011) Sustainable medicines and global health care. Planta Med 77(11):1129–1138
56. Kingston DGI (2011) Modern natural products drug discovery and its relevance to biodiversity
conservation. J Nat Prod 74(3):496–511
57. Narula A, Kumar S, Bansal K., Srivastava P (2004) Biotechnological approaches towards
improvement of medicinal plants. In Plant biotechnology and molecular markers
58. Alam G, Peppelenbos L (2009) Cultivation of medicinal plants in Uttarakhand. Econ Polit
Wkly 99–104
788 K. Kapoor et al.
59. Ghayur MN, Gilani AH (2005) Gastrointestinal stimulatory and uterotonic activities of dietary
radish leaves extract are mediated through multiple pathways. Phytotherapy research: An
international journal devoted to pharmacological and toxicological evaluation of natural prod-
uct derivatives 19(9):750–755
60. Cragg GM, Katz F, Newman DJ, Rosenthal J (2012) The impact of the United Nations con-
vention on biological diversity on natural products research. Nat Prod Rep 29(12):1407–1423
61. Morrison C, Humphries F, Lawson C (2021) A regional review of genetic resource access and
benefit sharing – key issues and research gaps. Environ Policy Law 51:1–24
62. David B, Wolfender J-L, Dias DA (2015) The pharmaceutical industry and natural products:
historical status and new trends. Phytochem Rev 14(2):299–315
63. Koehn FE, Carter GT (2005) The evolving role of natural products in drug discovery. Nat Rev
Drug Discov 4(3):206–220
64. Newman DJ (2008) Natural products as leads to potential drugs: an old process or the new
Hope for drug discovery? J Med Chem 51(9):2589–2599
65. Evans BE, Rittle KE, Bock MG, DiPardo RM, Freidinger RM, Whitter WL et al (1988)
Methods for drug discovery: development of potent, selective, orally effective cholecystokinin
antagonists. J Med Chem 31(12):2235–2246
66. Duarte CD, Barreiro EJ, Fraga CA (2007) Privileged structures: a useful concept for the ratio-
nal design of new lead drug candidates. Mini Rev Med Chem 7(11):1108–1119
67. Mason JS, Morize I, Menard PR, Cheney DL, Hulme C, Labaudiniere RF (1999) New 4-point
pharmacophore method for molecular similarity and diversity applications: overview of the
method and applications, including a novel approach to the design of combinatorial libraries
containing privileged substructures. J Med Chem 42(17):3251–3264
68. Kinghorn AD, Chin Y-W, Swanson SM (2009) Discovery of natural product anticancer agents
from biodiverse organisms. Curr Opin Drug Discov Devel 12(2):189–196
69. Tsang CK, Qi H, Liu LF, Zheng XF (2007) Targeting mammalian target of rapamycin (mTOR)
for health and diseases. Drug Discov Today 12(3-4):112–124
Biochanin A Chemistry, Structural
Modifications, and Therapeutic
Applications: An Update
Potential drug leads have come primarily from natural sources. There has been a
drop in their use in drug discovery and development in recent years, although while
combinatorial chemistry provides a wide range of structures, natural products offer
a unique opportunity to identify new low-molecular weight lead molecules [1–7].
Since less than 10% of the global biodiversity has been investigated for biological
activity, there are many more useful natural lead compounds to be discovered. The
problem is in obtaining this natural chemical diversity [4]. There is an infinite sup-
ply of creative scaffolds for developing effective medications from natural
Adil Farooq Wali and Roohi Mohi-ud-din contributed equally with all other contributors.
M. Maqbool
Pharmacy Practice Division, Department of Pharmaceutical Sciences, University of Kashmir,
Srinagar, Kashmir, India
K. Shenmar · R. H. Mir
Pharmaceutical Chemistry Division, Chandigarh College of Pharmacy,
Landran, Punjab, India
A. Akther
School of Health Science, University of Petroleum and Energy Studies,
Uttarakhand, Dehradun, India
A. F. Wali (*)
Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK
Medical and Health Science University, Ras Al Khaimah, UAE
e-mail: [email protected]
R. Mohi-ud-din (*)
Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS),
Srinagar, Jammu and Kashmir, India
Fig. 1 Biochanin A HO O
(5,7-dihydroxy-4′- A C
methoxy-isoflavone, BCA)
B
OH O
O
(1)
3 Chemistry of Biochanin A
One of the oldest known natural chemical scaffolds showing many biological activi-
ties is isoflavones. BCA is an O-methylated isoflavone with molecular formula
(C16H12O5) and molecular weight 284.267. Due to the 2,3-double bond in the ring C,
the –OH present at position 7 in ring A and the position-4′ in ring B, the present
isoflavone exhibits multifaceted bioactivity. Various derivitizations in these posi-
tions of significance have underlined their importance in the structure-activity rela-
tionships [35]. Chemically, its structure resembles estrogen, and it is thought to act
792 M. Maqbool et al.
on ERα and ERβ subtypes of estrogen receptors (ERs). It shows both agonistic and
antagonistic action on these receptors with ERβ functioning as a tumor suppressor
by opposing the mitogenic action of ERα [7] Physically, it is a white crystalline
powder with a melting point of 210–213 °C and solubility of 1 mg/mL in ethanol,
30 mg/mL in DMSO and DMF and 0.0583 mg/mL in water. Due to its poor aqueous
solubility, it has limited oral absorptivity. Various attempts have been made to
improve BCA’s bioavailability and solubility by using liposomes, dispersion agents,
silver nanoparticles, and synthetic manipulation [5].
In 2018, Gebreyohannes and Sherif synthesized genistein, BCA, and their analogs
(Fig. 2) to show the structure-activity relationship between the various substituents
present on rings A, B, and C and the DPPH (1,1-diphenyl-2-picrylhydrazyl) radical
scavenging activity. It was found that compound (2), which did not have any sub-
stituent on ring B, was the most potent analog exhibiting radical scavenging activity
of 86.95 ± 3.94% inhibition at 100 𝜇g/ml followed by genistein (84.89 ± 2.155). It
was postulated that whenever the –OH in ring B at position 4′ was masked by an
alkyl group, the compound became inactive. BCA backed this, and compound (3)
did not show any activity in the said assay. However, the study also showed that the
hydroxyl groups present at positions 5 and 7 of ring A played a bigger role in modu-
lating antioxidant activity as proven by moderate DDPH radical scavenging activity
of the other analogs (4) (74.77 ± 1.90), (5) (73.81 ± 1.61), (6) (76.36 ± 1.42) and (7)
(65.56 ± 1.15) [56].
Due to the isoflavone nucleus present in BCA, it also displays estrogenic activity.
However, to improve the poor oral bioavailability and aqueous solubility, Alexia
et al. synthesized ester and carbamate derivatives of BCA. All the 5 derivatives
synthesized along with BCA were tested for the estrogenic and antiproliferative
HO O
HO O HO O
OH O
OH O OH O
2 F NO2
3 4
HO O
HO O O O
OH O
CH3 OH O O O
5 Br Br
6 7
O O
O O O
O
N N
N
N N
NH2
9 10 11
activities by using human breast and endometrial adenocarcinoma cells. The results
showed that compound (8) (Fig. 3) displayed the maximum receptor-binding affin-
ity (RBA) with the relative ERα- and ERβ-binding affinity of 0.06 ± 0.01 and
0.08 ± 0.02. It was found that (8) affects MCF-7 and Ishikawa cells by induction of
gene expression to a level similar to 17β-estradiol and BCA but, contrary to the
action of both of the latter, for suppression of cell proliferation as well. In addition,
(8) appeared to display higher stability than all the other derivatives synthesized and
BCA in both MCF-7 and Ishikawa cells. The authors concluded that compound (8)
offers a safer alternative for hormone replacement therapy as compared to BCA [55].
Nontuberculous mycobacteria (NTM) are omnipresent bacteria that usually
cause infections when acquired through water, soil, animals, or artificial water res-
ervoirs like swimming pools, etc. They are generally grouped under the
Mycobacterium avium complex (MAC) comprising of M. intracellulare and
M. avium and are responsible for NTM infections in various patient populations
such as immunocompromised group, HIV positive, and cystic fibrosis. It was found
that BCA was an efflux pump inhibitor (EPI) of M. smegmatis, a related NTM spe-
cies. To synthesize EPIs active against MAC infections, the structure of BCA was
modified based on the exhaustive computational study on EPIs available in the lit-
erature. It was found that by getting rid of the phenolic –OH in ring A of BCA, by
isosteric replacement of O with NH, and by replacing the 4′- methoxy group in ring
B with propyloxy substituent, excellent MAC EPIs were obtained. The work carried
out by Cochiti et al.in 2017 reported the compound (9 10 and 11) belonging to the
modified 3-phenylquinolone scaffold obtained from BCA modification (Fig. 4)
were able to reverse the resistance of the M. avium strains to all the antimycobacte-
rial drugs and EtBr, further increasing their antimicrobial efficacies to record highs.
794 M. Maqbool et al.
5.1 Antihyperglycemic Effect
Pharmacological therapies for diabetes management have been linked to side effects
such as liver damage, lactic acidosis, diarrhea, and diminished responsiveness with
continuous use. Currently, the quest to discover natural substances capable of treat-
ing diabetes and other disorders has lasted decades, with biochanin A being men-
tioned as a possible candidate. In streptozotocin-induced diabetic rats, its
antihyperglycemic activity was examined. The scientists reported that after taking
biochanin A orally, normal plasma glucose, glycosylated hemoglobin, and insulin
levels were restored due to improved glycolytic enzyme activity [34, 49]. The
PPAR-α and PPAR-agonistic activities of natural compounds, such as BioA, further
supported their antidiabetic potential [34, 59].
5.2 Osteogenic Effect
Osteoporosis has been associated with an increase in adipogenesis in the bone mar-
row (osteoporosis due to age or menopause). There is evidence that isoflavones can
aid in the reduction of bone loss associated with aging or menopause [34, 60, 61].
Su et al. investigated the effects of biochanin A on osteoblast and adipocyte forma-
tion in a study using adipose-derived stem cells. Biochanin A was proven to reduce
adipocyte differentiation, PPAR-γ, and leptin and osteopontin mRNA expression,
Biochanin A Chemistry, Structural Modifications, and Therapeutic Applications… 795
HO O
Genistein+ BCA+
O
O
Formononetin (12)
HO O
HO O
SO3Na
OH O
OH O O
O
13 14
O COOCH 3
O O
O COOCH 3
H3COOC O
O
H3COOC OH
15
O
O
HO
O
O O O
O
Cr O
O O OH
HO
16
O
while osteogenic marker genes and osteocalcin expression were used to promote
alkaline phosphatase activity and mineralization. Additionally, transcription factor
2, osteoprotegerin, and Ras homolog family member A (Rho A) were activated.
This indicates that biochanin A promotes bone development [34, 62]. On the other
hand, biochanin A has been shown to prevent bone loss in ovariectomized rats at
796 M. Maqbool et al.
5.3 Gastroprotective Effect
Gastric ulceration is connected with oxidative stress and the formation of free radi-
cals. Preclinical studies indicate that flavonoids may protect against ethanol-induced
stomach ulcers by lowering excessive reactive oxygen species generation [34, 63].
Biochanin A has been shown to protect the stomach mucosa from damage caused by
ethanol in rats. In this study, enhanced nitric oxide and superoxide dismutase activ-
ity, decreased malondialdehyde levels, and, therefore, alleviated oxidative stress
[34, 64]. The gastroprotective activity of its derivative, genistein, was also revealed
in a study where it decreased indomethacin-induced gastropathy in rats by reducing
oxidative stress, decreasing inflammatory response, and repairing mucoprotective
functionality [34, 65].
5.4 Neuroprotective Activity
Aluminum, another neurotoxic toxin that stimulates glial cells, also produces
neuroinflammation through NF-κB signaling, leading to the production of prosta-
glandins, tumor necrosis factor-α, and nitric oxide. Aluminum buildup results in an
amyloid beta deposition in the brain, leading to neurodegeneration [34, 71, 72].
Recently, a biochanin-rich chickpea extract was established. This biochanin A rich
extract showed a protective role in aluminum-induced molecular neurodegeneration
by inhibiting NF-κB signaling [34, 73]. Biochanin A has previously been demon-
strated to suppress the NF-κB activation pathway by suppressing the degradation of
IκB in various experimental models [34, 74]. In scopolamine-treated aged mice,
MDA levels increased and decreased dramatically, respectively, due to the antioxi-
dant properties of the drug [34, 75].
Increased glutamate levels in cerebral ischemia cause neurotoxicity and cell
death. It has been proven that drugs intended to mitigate neurotoxicity following an
ischemic stroke should target the glutamate oxaloacetate transaminase enzyme,
which is implicated in glutamate metabolism. In this context, biochanin A has been
shown to be a potent stimulator of glutamate oxaloacetate transaminase production
and activity in brain cells. It markedly boosted mRNA and protein expression of
glutamate oxaloacetate transaminase and safeguarded cells against glutamate-
induced mortality [34, 76]. However, when the p75 neurotrophin death receptor
(p75NTR) is overexpressed, it has been linked to the death of neurons exacerbated
by peripheral neuropathy or spinal cord injury. Biochanin A, which blocks p38-
MAPK, was very good at reducing ibuprofen-induced p75NTR expression and
increasing cell survival in vitro [34, 77].
Several reports have revealed that biochanin A can negatively target the NF-κB
pathway with further inhibition of proinflammatory cytokines release, as NF-κB is
actively engaged in cytokine production. The mechanism involved could be phos-
phorylation activation of Iκβα, which is the inhibitor of NF-κB. In addition, other
inflammatory pathways such as iNOS, P38 MAPK, and AFT-2 can also be regulated
by biochanin A through phosphorylation inhibition [83], thus, creating an anti-
inflammatory and antiproliferative scenario as well [79]. Indirectly, the JAK-STAT
pathway of NF-κB can also be targeted by biochanin A, initiating an anti-
inflammatory cascade [84]. Furthermore, biochanin A mediates the stoppage of
cytokine bombardment and other proinflammatory molecules such as TNF-α, IL-1β,
iNOS, and COX-2 through targeting microglial activation, thus declining cytokine
storm as microglia are often considered to be accountable for the cytokine-mediated
inflammatory cascade [67, 85]. In a previous study to treat an inflammatory condi-
tion, the total inflammatory cells such as neutrophils, eosinophils, and cytokines
were found to be suppressed by biochanin A [86]. This could further be the rationale
behind biochanin A-induced controlled inflammation. Therefore, inflammatory dis-
eases such as rheumatoid arthritis, gout, and asthma can be benefitted from bio-
chanin A. Furthermore, numerous neurodegenerative diseases like Alzheimer’s
disease, Parkinson’s diseases, Huntington’s chorea, amyotrophic lateral sclerosis,
and cerebral ischemia have extensively been characterized by neuroinflammation in
different brain regions, hence considering potent anti-inflammatory action of bio-
chanin A, it can be the possible moiety for the therapeutic interventions in theses
brain diseases.
Traditionally, natural products, including flavones, have been utilized to cure micro-
bial infections. First time in the year 1996, biochanin A was found to have antiviral
potential against herpesvirus 6 where it acted by downregulating the tyrosine kinase
phosphorylation [87]. Likewise, owing to the action of biochanin A on the NF-κB
transcription factor and regulation of Akt and ERK1/2 signaling molecules, it can
possess antimicrobial activity. In this context, biochanin A has been shown to
Biochanin A Chemistry, Structural Modifications, and Therapeutic Applications… 799
Since liver dysfunction and hepatoxicity can arise due to several factors, including
ROS and inflammation. Hence, the antioxidant and anti-inflammatory properties of
biochanin A can be the relieving criteria against liver damage [93]. In an animal
model, drug-induced hepatotoxicity was shown to be alleviated with the help of
biochanin A by modulating NF-κB/pAKT pathway. Bax3, caspase-3, and eNOS
activation were also found to be downregulated, keeping the liver cells apoptosis
minimal [94]. Other cascades like activated antioxidant molecule Nrf2/HO-1 and
inhibited inflammatory marker NLRP3 inflammasome has been demonstrated to be
initiated with the biochanin A treatment, thereby reducing the occurrence of acute
liver injury [95, 96]. Many chemically induced liver toxicity such as carbon tetra-
chloride, arsenic, lipopolysaccharide, and can be relieved when biochanin A was
administered as a hepatoprotective therapeutic agent. Here, the mechanisms
involved could be anti-inflammatory, antioxidant, and immunomodulatory actions
[93, 97]. In a nutshell, biochanin A can be either tested and used pre-clinically or
clinically for several liver pathogeneses.
6 Conclusion
Natural bioactive chemicals are currently gaining popularity due to their pharmaco-
logical potential. Biochanin A‘s anticancer and neuroprotective capabilities have
been intensively investigated in recent years. Biochanin A’s therapeutic role and
multimechanistic approach against many diseases have been disclosed in numerous
investigations. Biochanin A‘s anticancer, antioxidant, anti-inflammatory, antimicro-
bial, and neuroprotective activities have been discussed in this article. Despite the
800 M. Maqbool et al.
fact that it has been used in clinical trials for postmenopausal symptoms, more clini-
cal trials are needed to assess its efficacy and safety in other therapeutic uses.
Organic chemists could investigate the structure–activity connection of biochanin A
to create more bioavailable biochanin A derivative. Biochanin A toxicological pro-
filing, such as hepatotoxicity and nephrotoxicity, should also be investigated. It is
possible that biochanin A could serve as a unique and prospective lead isoflavone
for therapeutic development based on preliminary research. In addition, major
research efforts must be made to uncover the hidden potential and unknown mecha-
nisms of this dietary phytoestrogen.
References
1. Jan R, Shah AJ, Wani TU, Farooq S, Jachak SM, Masoodi MH (2021) Curry leaf: An insight
into its pharmacological activities, medicinal profile, and phytochemistry. 4:145–168
2. Mishra BB, Tiwari VK (2011) Natural products: an evolving role in future drug discovery. Eur
J Med Chem 46(10):4769–4807
3. Rey-Ladino J, Ross AG, Cripps AW, McManus DP, Quinn R (2011) Natural products and the
search for novel vaccine adjuvants. Vaccine 29(38):6464–6471
4. Cragg GM, Newman DJ (2005) Biodiversity: a continuing source of novel drug leads. Pure
Appl Chem 77(1):7–24
5. Haefner B (2003) Drugs from the deep: marine natural products as drug candidates. Drug
Discov Today 8(12):536–544
6. Butler MS (2004) The role of natural product chemistry in drug discovery. J Nat Prod
67(12):2141–2153
7. Mir RH, Wani TU, Jan R, Shah AJ, Sabreen S, Mir PA, Rasool S, Masoodi MH, Bhat ZA
(2022) Nigella sativa as a therapeutic candidate for arthritis and related disorders. In: Black
seeds (Nigella Sativa). Elsevier, pp 295–312
8. Veeresham C (2012) Natural products derived from plants as a source of drugs. J Adv Pharm
Technol Res 3(4):200
9. Mohi-Ud-Din R, Mir RH, Wani TU, Shah AJ, Mohi-Ud-Din I, Dar MA, Pottoo FH (2021)
Novel drug delivery system for curcumin: implementation to improve therapeutic efficacy
against neurological disorders. Comb Chem High Throughput Screen 25:607–615
10. Mohi-Ud-Din R, Mir RH, Wani TU, Shah AJ, Banday N, Pottoo FH (2021) Berberine in the
treatment of neurodegenerative diseases and nanotechnology enabled targeted delivery. Comb
Chem High Throughput Screen 25:616–633
11. Mohi-Ud-Din R, Mir RH, Mir PA, Farooq S, Raza SN, Raja WY, Masoodi MH, Singh IP, Bhat
ZA (2021) Ethnomedicinal uses, Phytochemistry and pharmacological aspects of the genus
Berberis Linn: a comprehensive review. Comb Chem High Throughput Screen 24(5):624–644
12. Hong J (2014) Natural product synthesis at the interface of chemistry and biology. Chemistry
(Weinheim an der Bergstrasse, Germany) 20(33):10204
13. Zafar M, Sarfraz I, Rasul A, Jabeen F, Samiullah K, Hussain G, Riaz A, Ali M (2018)
Tubeimoside-1, triterpenoid saponin, as a potential natural cancer killer. Nat Prod Commun
13(5):1934578X1801300530
14. Mohi-Ud-Din R, Mir RH, Shah AJ, Sabreen S, Wani TU, Masoodi MH, Akkol EK, Bhat ZA,
Khan H (2021) Plant-derived natural compounds for the treatment of amyotrophic lateral sclero-
sis: an update. Curr Neuropharmacol. https://doi.org/10.2174/1570159X19666210428120514
15. Shah AJ, Mir RH, Mohi-Ud-Din R, Pottoo FH, Masoodi MH, Bhat ZA (2021) Depression:
an insight into heterocyclic and cyclic hydrocarbon compounds inspired from natural sources.
Curr Neuropharmacol 19(11):2020–2037
Biochanin A Chemistry, Structural Modifications, and Therapeutic Applications… 801
57. Cannalire R, Machado D, Felicetti T, Costa SS, Massari S, Manfroni G, Barreca ML, Tabarrini
O, Couto I, Viveiros MJ (2017) Natural isoflavone Biochanin A as a template for the design of
new and potent 3-phenylquinolone efflux inhibitors against Mycobacterium avium. Eur J Med
Chem 140:321–330
58. Dong Q, Wang D, Li L, Wang J, Li Q, Duan L, Yin H, Wang X, Liu Y, Yuan G (2022) Biochanin
A sensitizes glioblastoma to Temozolomide by inhibiting autophagy. Mol Neurobiol:1–11
59. Sundaresan A, Radhiga T, Deivasigamani B (2018) Biological activity of Biochanin A: a
review. Asian J Pharm Pharmacol 4(1):1–5
60. Atkinson C, Compston JE, Day NE, Dowsett M, Bingham SA (2004) The effects of phytoes-
trogen isoflavones on bone density in women: a double-blind, randomized, placebo-controlled
trial. Am J Clin Nutr 79(2):326–333
61. Kawakita S, Marotta F, Naito Y, Gumaste U, Jain S, Tsuchiya J, Minelli E (2009) Effect of an
isoflavones-containing red clover preparation and alkaline supplementation on bone metabo-
lism in ovariectomized rats. Clin Interv Aging 4:91
62. Su S-J, Yeh Y-T, Su S-H, Chang K-L, Shyu H-W, Chen K-M, Yeh H (2013) Biochanin A
promotes osteogenic but inhibits adipogenic differentiation: evidence with primary adipose-
derived stem cells. Evid Based Complement Alternat Med 2013
63. Royer M, Diouf PN, Stevanovic T (2011) Polyphenol contents and radical scavenging capaci-
ties of red maple (Acer rubrum L.) extracts. Food Chem Toxicol 49(9):2180–2188
64. Hajrezaie M, Salehen N, Karimian H, Zahedifard M, Shams K, Batran RA, Majid NA, Khalifa
SA, Ali HM, El-Seedi H (2015) Biochanin A gastroprotective effects in ethanol-induced gas-
tric mucosal ulceration in rats. PLoS One 10(3):e0121529
65. Vivatvakin S, Werawatganon D, Somanawat K, Klaikeaw N, Siriviriyakul P (2017) Genistein-
attenuated gastric injury on indomethacin-induced gastropathy in rats. Pharmacogn Mag
13(Suppl 2):S306
66. Duty S, Jenner P (2011) Animal models of Parkinson’s disease: a source of novel treatments
and clues to the cause of the disease. Br J Pharmacol 164(4):1357–1391
67. Wang J, Wu W-Y, Huang H, Li W-Z, Chen H-Q, Yin Y-Y (2016) Biochanin A protects against
lipopolysaccharide-induced damage of dopaminergic neurons both in vivo and in vitro via
inhibition of microglial activation. Neurotox Res 30(3):486–498
68. Sanders LH, Greenamyre JT (2013) Oxidative damage to macromolecules in human Parkinson
disease and the rotenone model. Free Radic Biol Med 62:111–120
69. Chen H, Wang X, Wang M, Yang L, Yan Z, Zhang Y, Liu Z (2015) Behavioral and neurochemi-
cal deficits in aging rats with increased neonatal iron intake: silibinin’s neuroprotection by
maintaining redox balance. Front Aging Neurosci 7:206
70. Yu L, Wang X, Chen H, Yan Z, Wang M, Li Y (2017) Neurochemical and behavior deficits
in rats with iron and rotenone co-treatment: role of redox imbalance and neuroprotection by
Biochanin A. Front Neurosci 11:657
71. Bondy SC (2014) Prolonged exposure to low levels of aluminum leads to changes associated
with brain aging and neurodegeneration. Toxicology 315:1–7
72. Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F (2012) New pharmacological strat-
egies for treatment of Alzheimer’s disease: focus on disease modifying drugs. Br J Clin
Pharmacol 73(4):504–517
73. Wahby M, Mohammed D, Newairy A, Abdou H, Zaky A (2017) Aluminum-induced molecular
neurodegeneration: the protective role of genistein and chickpea extract. Food Chem Toxicol
107:57–67
74. Wu D-Q, Zhong H-M, Ding Q-H, Ba L (2014) Protective effects of Biochanin A on articular
cartilage: in vitro and in vivo studies. BMC Complement Altern Med 14(1):1–10
75. Biradar S, Joshi H, Chheda T (2014) Biochanin-A ameliorates behavioural and neurochemical
derangements in cognitive-deficit mice for the betterment of Alzheimer’s disease. Hum Exp
Toxicol 33(4):369–382
804 M. Maqbool et al.
76. Khanna S, Stewart R, Gnyawali S, Harris H, Balch M, Spieldenner J, Sen CK, Rink C (2017)
Phytoestrogen isoflavone intervention to engage the neuroprotective effect of glutamate oxalo-
acetate transaminase against stroke. FASEB J 31(10):4533–4544
77. Kalb R (2005) The protean actions of neurotrophins and their receptors on the life and death
of neurons. Trends Neurosci 28(1):5–11
78. Xiao P, Zheng B, Sun J, Yang J (2017) Biochanin A induces anticancer effects in SK-Mel-28
human malignant melanoma cells via induction of apoptosis, inhibition of cell invasion and
modulation of NF-κB and MAPK signaling pathways. Oncol Lett 14(5):5989–5993
79. Hsu Y-N, Shyu H-W, Hu T-W, Yeh J-P, Lin Y-W, Lee L-Y, Yeh Y-T, Dai H-Y, Perng D-S,
Su S-H (2018) Anti-proliferative activity of Biochanin A in human osteosarcoma cells via
mitochondrial-involved apoptosis. Food Chem Toxicol 112:194–204
80. Wang Y, Gho WM, Chan FL, Chen S, Leung LK (2008) The red clover (Trifolium pratense)
isoflavone Biochanin A inhibits aromatase activity and expression. Br J Nutr 99(2):303–310
81. Cho I-A, You S-J, Kang K-R, Kim S-G, Oh J-S, You J-S, Lee G-J, Seo Y-S, Kim DK, Kim
CS (2017) Biochanin-A induces apoptosis and suppresses migration in FaDu human pharynx
squamous carcinoma cells. Oncol Rep 38(5):2985–2992
82. Youssef MM, Tolba MF, Badawy NN, Liu AW, El-Ahwany E, Khalifa AE, Zada S, Abdel-
Naim AB (2016) Novel combination of sorafenib and biochanin-A synergistically enhances
the anti-proliferative and pro-apoptotic effects on hepatocellular carcinoma cells. Sci Rep
6(1):1–12
83. Kole L, Giri B, Manna SK, Pal B, Ghosh S (2011) Biochanin-A, an isoflavon, showed anti-
proliferative and anti-inflammatory activities through the inhibition of iNOS expression, p38-
MAPK and ATF-2 phosphorylation and blocking NFκB nuclear translocation. Eur J Pharmacol
653(1–3):8–15
84. Basu A, Das AS, Borah PK, Duary RK, Mukhopadhyay R (2020) Biochanin A impedes STAT3
activation by upregulating p38δ MAPK phosphorylation in IL-6-stimulated macrophages.
Inflamm Res 69(11):1143–1156
85. Chen H-Q, Jin Z-Y, Li G-H (2007) Biochanin A protects dopaminergic neurons against
lipopolysaccharide-induced damage through inhibition of microglia activation and proinflam-
matory factors generation. Neurosci Lett 417(2):112–117
86. Ko W-C, Lin L-H, Shen H-Y, Lai C-Y, Chen C-M, Shih C-H (2011) Biochanin A, a phytoes-
trogenic isoflavone with selective inhibition of phosphodiesterase 4, suppresses ovalbumin-
induced airway hyperresponsiveness. Evid Based Complement Alternat Med 2011
87. Cirone M, Zompetta C, Tarasi D, Frati L, Faggioni A (1996) Infection of human T lymphoid
cells by human herpesvirus 6 is blocked by two unrelated protein tyrosine kinase inhibitors,
Biochanin A and herbimycin. AIDS Res Hum Retrovir 12(17):1629–1634
88. Sithisarn P, Michaelis M, Schubert-Zsilavecz M, Cinatl J Jr (2013) Differential antiviral and
anti-inflammatory mechanisms of the flavonoids Biochanin A and baicalein in H5N1 influenza
A virus-infected cells. Antivir Res 97(1):41–48
89. Michaelis M, Sithisarn P, Cinatl J Jr (2014) Effects of flavonoid-induced oxidative stress on
anti-H5N1 influenza a virus activity exerted by baicalein and Biochanin A. BMC Res Notes
7(1):1–6
90. Hanski L, Genina N, Uvell H, Malinovskaja K, Gylfe Å, Laaksonen T, Kolakovic R, Mäkilä
E, Salonen J, Hirvonen J (2014) Inhibitory activity of the isoflavone Biochanin A on intracel-
lular bacteria of genus chlamydia and initial development of a buccal formulation. PLoS One
9(12):e115115
91. Zhao X, Tang X, Guo N, An Y, Chen X, Shi C, Wang C, Li Y, Li S, Xu H (2018) Biochanin
A enhances the defense against salmonella enterica infection through AMPK/ULK1/mTOR-
mediated autophagy and extracellular traps and reversing SPI-1-dependent macrophage (MΦ)
M2 polarization. Front Cell Infect Microbiol 8:318
92. Ross R, Conti P (2020) COVID-19 induced by SARS-CoV-2 causes Kawasaki-like disease
in children: role of pro-inflammatory and anti-inflammatory cytokines. J Biol Regul Homeost
Agents 34:767–773
Biochanin A Chemistry, Structural Modifications, and Therapeutic Applications… 805
93. Breikaa RM, Algandaby MM, El-Demerdash E, Abdel-Naim AB (2013) Biochanin A protects
against acute carbon tetrachloride-induced hepatotoxicity in rats. Biosci Biotechnol Biochem
77(5):909–916
94. Chaturvedi S, Malik MY, Azmi L, Shukla I, Naseem Z, Rao C, Agarwal NK (2018)
Formononetin and Biochanin A protects against ritonavir induced hepatotoxicity via modula-
tion of NfκB/pAkt signaling molecules. Life Sci 213:174–182
95. Breikaa RM, Algandaby MM, El-Demerdash E, Abdel-Naim AB (2013) Multimechanistic
antifibrotic effect of Biochanin A in rats: implications of proinflammatory and profibrogenic
mediators. PLoS One 8(7):e69276
96. Liu X, Wang T, Liu X, Cai L, Qi J, Zhang P, Li Y (2016) Biochanin A protects
lipopolysaccharide/D-galactosamine-induced acute liver injury in mice by activating the Nrf2
pathway and inhibiting NLRP3 inflammasome activation. Int Immunopharmacol 38:324–331
97. Jalaludeen AM, Ha WT, Lee R, Kim JH, Do JT, Park C, Heo YT, Lee WY, Song H (2016)
Biochanin A ameliorates arsenic-induced hepato-and hematotoxicity in rats. Molecules
21(1):69
Dietary Natural Polyphenols Against
Bacterial and Fungal Infections:
An Emerging Gravity in Health Care
and Food Industry
1 Introduction
Polyphenols are naturally available plant composites and also the most significant
antioxidant for human beings. The gastrointestinal tract is the essential organ gave
to consume less calories segments, the eating routine might be viewed as one of the
fundamental variables in the usefulness, respectability and creation of digestive
microbiota [1]. In the gastrointestinal tract, numerous polyphenols remain unab-
sorbed in the digestive organ, where the gastrointestinal microbiota are generally
processed. While expecting essential jobs for progressing host prosperity, this
digestive well-being climate is introduced with the impact of outer impacts,
B. Patra
School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla, Sambalpur, Odisha, India
N. Das
Department of Botany, University of Science and Technology Meghalaya, Techno City,
Baridua, Ri-Bhoi, Meghalaya, India
M. Z. Shamim
Department of Food Nutrition and Dietetics, Faculty of Sciences, Assam Down Town
University, Gandhinagar, Panikhaiti, Guwahati, Assam, India
T. K. Mohanta
Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
B. Mishra
Department of Biotechnology, Chaitanya Bharathi Institute of Technology (CBIT), Gandipet,
Hyderabad, Telangana, India
Y. K. Mohanta (*)
Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied
Biology, University of Science and Technology Meghalaya, Techno City, Baridua, Ri-Bhoi,
Meghalaya, India
e-mail: [email protected]
including dietary patterns [2]. A couple procedures have been created to build dis-
solvability and transport across the gastrointestinal parcel and move it to designated
digestive districts to determine dietary polyphenols at the low bioavailability.
Dietary polyphenols are normal plant-based mixtures, including food sources, for
example, vegetables, grains, organic products, espresso, tea, wine, etc. [3].
Hydroxylated phenyl moieties characterize polyphenols as only a huge heteroge-
neous gathering of mixtures. Polyphenols are typically sorted into flavonoids and
non-flavonoids in view of their compound construction and intricacy (counting phe-
nolic rings and replacement gatherings). Human body sees polyphenols as xenobi-
otics, and their bioavailability is respectably poor [4]. Recently, more than 8000
polyphenols have been distinguished and are grouped by their carbon skeleton idea:
flavonoids, phenolic acids, lignans and stilbenes. The organization, just as the pro-
portion of various species that structure the digestive microbiome, is exceptionally
different inside the human populace, and every individual has their own interesting
profile of microbial species, which can measure up to a fingerprint [5]. The separa-
tion of stomach microbiota structure and profile is brought about by the impact of
various and different elements, like age, beginning, geological area, climate, dietary
propensities, probiotics, health and the utilization of anti-infection agents [6].
Commensal microbes produce bacteriocins that explicitly repress individuals from
the equivalent or comparable bacterial species (e.g. E. coli versus microorganism
enterohemorrhagic E. coli). Commensal microbes produce short-chain unsaturated
fats and cause pH decrease, subsequently forestalling the colonization by microor-
ganisms whose ideal pH for development is natural [7]. The extraordinary variety of
bacterial species shaping the stomach microbiota ensnares the huge number of qual-
ities which they contain and the huge metabolic limit of the gastrointestinal micro-
biome, which is roughly 100-crease more prominent than that of the human liver
[8]. Digestive microbes add to the breakdown of polysaccharides and polyphenols
just as partake in the synthesis of nutrients (B12, K).
1.1 Sources of Polyphenols
tea, implantations and so forth), while others are explicit to specific food varieties
(flavanones in citrus organic product, isoflavones in soya and phloridzin in apples).
Much of the time, food varieties contain complex combinations of polyphenols,
which are regularly inadequately characterized [11]. Apples contain flavanol mono-
mers and oligomers, chlorogenic corrosive and little amounts of other hydroxycin-
namic acids, 2 glycosides of phloretin, a few quercetin glycosides and anthocyanins
and cyanidin 3-galactoside in the skin of specific varieties [12]. Apples are one of
the uncommon kinds of nourishment for which genuinely exact information on
polyphenol composition are accessible.
Flavours has large history of being utilized as normal food additives with people
medication. This is a direct result of antimicrobial impact which contains saponin,
alkaloids, flavonoids, anthocyanin, phenolic acids and tannins [13]. Rejuvenating
balm from polyphenols is enormous gatherings of organization with most grounded
antimicrobial impact. Many examination distributions detailing in various kinds of
plant arrangements can apply both positive and adverse consequence on organisms.
Taking into account that polyphenols have been demonstrated to build the develop-
ment of Akkermansia muciniphila, the valuable effect of polyphenols on human
well-being might result from other than cancer prevention activity [14]. Every one
of these explorations recommend that polyphenols arriving at the digestive organ
may not exclusively be catabolized to phenolic acids additionally impacts of gastro-
intestinal probiotic microorganisms. Taken together, polyphenols have all the ear-
marks of being ready to adjust stomach microecology and, by influencing the
absolute number of advantageous species in the stomach, may give positive stom-
ach health [15]. The impact of pure polyphenols and plant extract, just as the
strength of that effect on microscopic organisms, contrasts relying upon the sort of
both phenolic mixtures and microbial strain. The antibacterial movement of flavo-
noids might result from their capacity to frame complexes with proteins through
certain forces, for example, hydrogen holding and hydrophobic impact just as by
covalent bond formation [12]. Proteins adjusted by polyphenols restricting have a
certain amino acid obstructed and go through conformity advances, which can
cause changes in protein structure, hydrophobicity, dissolvability and isoelectric
point. In phenolic protein complexation, prompt changes in their physicochemical
and organic properties including the absorbability and use of food proteins as well
as digestive enzyme activities [14]. Quinones are recognized to composite irrevers-
ibly with nucleophilic amino acids in protein compounds, which prompts inactiva-
tion and loss of capacity in the proteins. They potentially associate with cell divider
polypeptides, layer-bound catalysts and surface-uncovered adhesins of pathogenic
bacteria [15]. The catalysts cysteine transpeptidases are utilized by Gram-positive
bacteria to show proteins in the surface of the cell glycoproteins, and they can con-
nect to proteins in the cross-span peptide of the cell divider. The inhibitory effect of
810 B. Patra et al.
polyphenols on the activity of the bacterial efflux siphon, which changes transport
through the cell divider and cytoplasmic layers are additionally considered.
Flavonoids can modulate the action of bacterial proteins, which are significant for
cell life [16]. The union of nucleic corrosive can be hindered by polyphenols like-
wise through topoisomerase restraint. Flavonoids are inhibitors of topoisomerases,
and it assumes a significant part in their antimycobacterial activity [17]. The signifi-
cant capacity of the cell wall is to give cell integrity and shape to act as an osmotic
obstruction. Gram-negative microbes were resistant for numerous antibacterial sub-
stances because of the hydrophilic surface of their external layer and related pro-
teins in the periplasmic space, which are equipped for separating numerous
atoms [18].
neurodegenerative infections. Some data are accessible on the qualities and quanti-
ties of polyphenols that are consumed every day all throughout the world [12]. This
information has been acquired through examination of the principle of hydrolysis of
glycosides and esters in the food varieties. Utilization of these substances has been
assessed at 20–25 mg/d in the Denmark, Holland and United States. In Spain, the
complete utilization of catechins and proanthocyanin dimers has been assessed at
18–31 mg per day, and the primary sources are pears, apples, red wine and grapes.
Utilization of significant constituents might differ profoundly indicated by tea and
coffee consumption. A few people who drink a few cups each day might consume
as much as 500–800 mg hydroxycinnamic acids per day [13]. Regulation of cell
signalling pathways by polyphenols may help essentially to clarify the components
of the activities of polyphenol-rich compounds. Some studies have additionally
shown that different polyphenol subgroups might vary essentially in stability, bio-
availability and physiological capacities connected with human health [14].
Large numbers of phenolic structures are presently known, and among them more
than 4000 flavonoids have been distinguished. Polyphenols also artificially por-
trayed as mixtures with phenolic constituents. This group natural products are
exceptionally varied and contains few sub-groups of phenolic components. Organic
products, vegetables and different sorts of food sources like chocolate, tea and wine
are rich source of polyphenols [15]. The variety and wide dissemination of polyphe-
nols in plants have prompted various approaches to arranging these normally hap-
pening compounds. Polyphenols have been grouped by their natural capacity, source
of origin and chemical construction. Phenolic acids are non-flavonoid polyphenolic
intensifies that can be additionally separated into two principle types, benzoic cor-
rosive and cinnamic corrosive [16]. Flavonoids have the common structure in which
the two C6 units of ring A and ring B are of phenolic characteristics. The fundamen-
tal designs of flavonoids are aglycones. In plants, the majority of these mixtures
exist as glycosides. Organic exercises of these mixtures, including cell reinforce-
ment action, rely upon both the underlying contrast and the glycosylation patterns
[18]. They are generally found in the leguminous group in plants. Since beans, espe-
cially soybean, are a significant eating regimen in many societies. Genistein and
daidzein are the two principle isoflavones found in soy [19]. Flavones and their
3-hydroxy subsidiaries flavonols including their glycosides methoxides of the rings
make the biggest subgroup among all polyphenols. The most well-known flavonol
aglycones, quercetin and kaempferol alone have not less than 279 and 347 unique
glycosidic blends, respectively [20]. Some flavanones have interesting substitution
designs, i.e. furanoflavanones, prenylated flavanones, pyranoflavanones and ben-
zylated flavanones, giving subordinates of this subgroup [21]. A notable flavanonol
is taxifolin from citrus organic products. Proanthocyanidins are generally viewed as
consolidated tannins. Anthocyanidins are the important parts of the red, blue and
812 B. Patra et al.
purple colours of most of bloom petals, vegetables and certain varieties of rice
grains. Anthocyanidins in plants mostly exist in glycosidic structures, which are
available as anthocyanins [22]. Delphinidin Cyanidin and Pelargonidin are the most
broadly found anthocyanidins with multiple monomeric anthocyanidins. Some
polyphenols may have N-containing utilitarian substituents. Two such gatherings of
polyphenolic amides are of importance for normal food source. Capsaicin are
accountable for the hotness of the chili peppers but found as cancer prevention agent
and mitigating properties and also oxidative protection context in cells [23]. The
phenolic acids, phenolic amides and flavonoids are a few non-flavonoid polyphe-
nols found in food varieties that are viewed as vital to human health. Among these,
resveratrol is interesting to the grapes and red wine and its subsidiaries are found in
berry organic products. In strawberries and in the skins of various berries nuts, these
ellagic acid derivatives are also available [24].
2.2 Microbial Sources
Diet assumes a prevailing part in tweaking the digestive microbiota. Large numbers
of these dietary substrates are prebiotics, which are specifically used by colonic
microorganisms giving medical advantages. The stomach microbiota is mostly
addressed by set of bacterial phyla, i.e. Actinobacteria, Firmicutes, Proteobacteria,
Verrucomicrobia and Bacteroidota [25]. High inter-individual variability, Firmicutes
is the biggest phylum in rodents with more than 250 genera. The phylum
Actinobacteria is less reliably distinguished as predominant and addresses a lower
rate of the absolute microscopic organisms. It incorporates the variety Bifidobacterium
from which numerous probiotics are collected [26]. Additionally, the phylum
Verrucomicrobia incorporates the variety Akkermansia spp., which is available in
low extent and is presently viewed as a next edge probiotic [27]. Other applicable
microorganisms considered as next-generation probiotics like A. muciniphila,
B. thetaiotaomicron and F. prausnitzii are interacting with the host immune system
and have been related with valuable impacts on the host [28].
Phenolic acids like gallic and cinnamic acids are viewed as metabolites of the shiki-
mate pathway. Biosynthesis of complex polyphenols, like flavonoids, is connected
to plastid and mitochondrial determined intermediates and each expecting com-
modity to the cytoplasm where they are consolidated into discrete pieces of the
molecule [29]. The aromatic compound is considered to start from the amino acid
phenylalanine and its result of the shikimate pathway considered as three units of
Dietary Natural Polyphenols Against Bacterial and Fungal Infections: An Emerging… 813
Polyphenols that are not digested in the intestine arrive at the colon. The microflora
hydrolyses glycosides into aglycones and broadly processes the aglycones into dif-
ferent good smelling acids [32]. Aglycones are parted by the launch of the hetero-
cycle at various structure relying upon their compound construction. Flavanols
primarily produce hydroxy phenylacetic acids, flavones and flavanones fundamen-
tally produce hydroxyphenyl propionic acids and flavanols principally produce
phenylvalerolactones and hydroxyphenyl propionic acids [33]. These acids are
additionally processed to subsidiaries of benzoic corrosive. The microbial metabo-
lites are consumed and formed with glycine, glucuronic corrosive or sulphate. The
cleavage and metabolic pathways are grounded in creatures, and the impact of sub-
stance structure on debasement is known [34]. However, some limited research data
are available to recognize the new microbial composition and metabolite produc-
tion. Interindividual varieties and the impact of the microflora structure and of the
typical eating regimen on microbial metabolite creation must be assessed. Late
investigations have shown that plasma fixations and urinary discharge of microbial
metabolites in people can be higher than those of tissue systems related to polyphe-
nols. For example, wine polyphenols that are not effectively absorbed in urinary
excretion of microbial metabolism in human health [35].
3.1 Antibacterial Activity
openings on the outer layer of the cell were seen, and even lysis was possible [28].
Other antimicrobial parts in the nano-encapsulated structures could be significantly
more productive against microorganisms. For instance, normal antimicrobials
showed better antimicrobial action when they were as nanosized particles.
β-Glucosidases are created because of glucoside-rich conditions by a few digestive
microbes like Blautia producta, Bifidobacterium spp., B. thetaiotaomicron, E. coli,
L. plantarum and Erysipelato clostridium ramosum [29]. Numerous lactic acid bac-
teria and some microscopic organisms (Streptococcus thermophilus, L. acidophilus,
Lactobacillus delbrueckii ssp. bulgaricus) are additionally ready to create
β-glucosidases [30]. Due to their well-being, these β-glucosidase-creating microor-
ganisms are utilized to build the bio accessibility of phenolic glucosides during
food aging.
3.2 Anti-candidal Activity
There has been a sharp ascent in the event of Candida diseases and related mortality
throughout the most recent couple of years because of the developing collection of
immunomodulated population. Limited accessible antifungal organisms, unwanted
side effects and toxicity represent a significant clinical test for the treatment of can-
didiasis [31]. However, components that got from regular sources displaying signifi-
cant antifungal properties are a promising hotspot for the advancement of novel
enemy of candidal treatment. Phenolic compounds disengaged from normal sources
have antifungal properties of interest [32]. Especially, phenolic acids have shown
promising in vitro and in vivo action against Candida species. Phenolic acid subor-
dinates separated from these sources, i.e. caffeic, gallic, benzoic, cinnamic, phenyl-
acetic acids and protocatechuic additionally have antifungal activity [33]. Therefore,
phenolic acids got from various normal sources have significant factor against
changeable MIC value of Candida species. Moreover, a potential impact on the
C. albicans has been displayed for caffeic acid subordinates which might interfere
1,3-β-glucan synthase. Some investigations have found metabolic pathways of phe-
nolic acids against Candida. In vitro immunoregulatory impact on monocytes
against C. albicans by cinnamic acid have eugenol inhibitory cell cycle at G1, S and
G2-M stages in C. albicans and subsequent instigates apoptosis [34]. Another phe-
nolic compound with association with curcumin additionally instigates apoptosis in
C. albicans, by expanding the responsive oxygen species (ROS) and acceptance of
CaMCA1 gene expression. In antifungal resistance, there are significant limits in
deficient spectrum range, less bioavailability with tolerance index, interaction with
different nanodrug medications, insufficient pharmacokinetic profile and extensive
poisonous effects [35]. Another review has shown that phenolic mixtures like thy-
mol and carvacrol fundamentally decline the articulation levels of harmfulness
qualities CDR1 and MDR1 in fluconazole-resistant C. albicans.
Dietary Natural Polyphenols Against Bacterial and Fungal Infections: An Emerging… 815
Smart packaging and developing demand for natural food sources are driving a
emergent interest in natural antimicrobials and effective opposing impact against a
wide scope of undesirable microorganisms in foods [39]. Controlling microbial
development in food items has forever been a major concern for the various partners
in the agricultural food sector. Some research should be thought for ensuring both
food safety and food waste deduction. Therefore, this peculiarity of antimicrobial
resistance and the presence of antibiotics effect build ups in the environment have
drawn in much consideration from present day consumers [40]. Thus, the antimicro-
bials action that are powerful against both pathogenic and waste microorganisms is
crucial. The presence of alkyl bunches in the fragrant core generates phenoxyl
extremists answered to improve the antibacterial efficacy of phenolics and change
their dissemination proportion between aqueous and non-fluid stages, including
bacterial phases [41]. The capacity to enter the cell film and interact with cell com-
ponents incites irreversible harm to the cell wall and coagulation of the cell content,
influencing both membrane and intracellular catalysts. Polyphenol dependability is
an essential property for application in food systems and is a component of a few
variables, such as chemical structure, size, water dissolvability and polarity [42].
However, nourishment has become a key factor to human health and maximal
knowledge of the impacts of treatment processes in fundamental for maintaining the
function of plant biomolecules not just as food preservatives but also builds of nutri-
tional concentration. The capacity of polyphenols for quite under specific environ-
mental conditions with high temperature and light could genuinely influence their
synthetic and physical stabilities [43]. Different phytochemicals and cancer preven-
tion activities purposefully introduced to food nutritional system might assist with
balancing polyphenols. Vitamin C added to processed yellow fruits natural product
816 B. Patra et al.
applied a defensive effect on plant chemicals [44]. Eatable coatings give an obstruc-
tion to vaporous exchange as well as the transmission of dampness, flavours and
other soluble constituents of processed items when stored for long long.
With the advantages of nanotechnology, the potential risk in food factory and their
application could be considered for health management. But the fact is there are no
proof on the health risk of polyphenol-stacked nanoparticles, the risk evaluation of
nanomaterials, nano silver, zinc oxide or silicon dioxide utilized for their exemplifi-
cation was reviewed [46]. The critical physicochemical properties of nanoparticles
that are answerable for toxicity including molecule size, surface coating and reac-
tivity, crystallography, total carrying capacities, surface coatings, combination and
readiness changes and sample purity [47]. The physicochemical properties of these
particles can establish a supportive effect on free radicals as well as cell death
inflammation. The biochemical movement of polyphenols is mostly connected with
their capacity to metals and free radical scavenge. However supportive oxidant con-
duct under specific conditions, prompting the arrangement of responsive oxygen
species that can harm lipids, DNA and other components [48]. Utilization of tea in
high dosages prompts an imbalance in the cancer prevention effect and favourable
to antioxidant activities of tea flavonoids, dietary iron absorption as well as intesti-
nal effect along with digestive enzyme from green tea. The decrease of gastrointes-
tinal assimilation of dietary iron and the precipitation of stomach related effect by
tannins from tea which has lipase action by polyphenols [49]. For the utilization of
phenolic compounds for food storage capacity, they must be stable until the termi-
nation of product which they were added [50]. Nonetheless, polyphenols are rela-
tively unstable when straight forwardly applied in food sources. The steadiness of
such compounds in food system can be credited to a progression of stabilities, phys-
iochemical compounds, colloidal and natural correlation with each other [51].
Dietary Natural Polyphenols Against Bacterial and Fungal Infections: An Emerging… 817
Acknowledgements: The authors would like to extend their sincere appreciation to their respec-
tive institutions for providing a suitable ambiance for developing this manuscript. Y.K.M. is highly
indebted and extends its sincere thanks to SERB-DST, Government of India for providing support
to his Nano-biotechnology and Translational Knowledge Laboratory through research Grant No.
SRG/2022/000641.
References
1. Ma B, Stepanov I, Hecht SS (2019) Recent studies on DNA adducts resulting from human
exposure to tobacco smoke. Toxics:7
2. Man S, Luo C, Yan M, Zhao G, Ma L, Gao W (2021) Treatment for liver cancer: from sorafenib
to natural products. Eur J Med Chem 224:113690
3. Sikiru AB, Arangasamy A, Egena SSA, Veerasamy S, Reddy IJ, Raghavendra B (2021)
Elucidation of the liver proteome in response to an antioxidant intake in rabbits. Egypt
Liver J:11
4. Izzo C, Annunziata M, Melara G, Sciorio R, Dallio M, Masarone M et al (2021) The
role of resveratrol in liver disease: a comprehensive review from in vitro to clinical trials.
Nutrients 13:1–23
5. Singh AK, Cabral C, Kumar R, Ganguly R, Rana HK, Gupta A et al (2019) Beneficial effects
of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency.
Nutrients 11
6. Milinčić DD, Popović DA, Lević SM, Kostić A, Tešić ŽL, Nedović VA et al (2019) Application
of polyphenol-loaded nanoparticles in food industry. Nano 9
7. Protease S-M, Case T, Panagiotopoulos AA, Karakasiliotis I, Kotzampasi D, Dimitriou M et al
(2021) Natural polyphenols inhibit the dimerization of the SARS-CoV-2 main protease: the
case of Fortunellin and its structural analogs. Molecules 26:6068
8. Pascoalino LA, Reis FS, Prieto MA, Barreira JCM, Ferreira ICFR, Barros L (2021) Valorization
of bio-residues from the processing of main portuguese fruit crops: from discarded waste to
health promoting compounds. Molecules:26
9. Tringali C (2020) Special issue: from natural polyphenols to synthetic bioactive analogues.
Molecules 25:3–5
818 B. Patra et al.
10. Ben-Othman S, Jõudu I, Bhat R (2020) Bioactives from agri-food wastes: present insights and
future challenges. Molecules 25:510
11. Xie Y, Chen J, Xiao A, Liu L (2017) Antibacterial activity of polyphenols: structure-activity
relationship and influence of hyperglycemic condition. Molecules:22
12. López-Yerena A, Domínguez-López I, Vallverdú-Queralt A, Pérez M, Jáuregui O, Escribano-
Ferrer E et al (2021) Metabolomics technologies for the identification and quantification of
dietary phenolic compound metabolites: an overview. Antioxidants. 10:1–25
13. de Falco B, Grauso L, Fiore A, Bochicchio R, Amato M, Lanzotti V (2021) Metabolomic
analysis and antioxidant activity of wild type and mutant chia (Salvia hispanica L.) stem and
flower grown under different irrigation regimes. J Sci Food Agric 101:6010–6019
14. Uriarte-fr G, Hern MM, Guti G, Santiago-ortiz MM, Morris-quevedo HJ, Meneses-mayo
M (2021) Pre-Hispanic foods oyster mushroom (Pleurotus ostreatus), Nopal (Opuntia ficus-
indica) and Amaranth (Amaranthus sp.) functional cookies. J Fungi 7:911
15. Elejalde E, Villarán MC, Alonso RM (2021) Grape polyphenols supplementation for exercise-
induced oxidative stress. J Int Soc Sports Nutr 18
16. Kumar A, Rani M, Mani S, Shah P, Singh DB, Kudapa H et al (2021) Nutritional significance
and antioxidant-mediated antiaging effects of finger millet: molecular insights and prospects.
Front Sustain Food Syst 5:1–17
17. Ofosu FK, Daliri EBM, Elahi F, Chelliah R, Lee BH, Oh DH (2020) New insights on the use
of polyphenols as natural preservatives and their emerging safety concerns. Front Sustain Food
Syst. https://doi.org/10.3389/fsufs.2020.525810
18. Cháirez-Ramírez MH, de la Cruz-López KG, García-Carrancá A (2021) Polyphenols as antitu-
mor agents targeting key players in cancer-driving signaling pathways. Front Pharmacol:1–25
19. Martinengo P, Arunachalam K, Shi C (2021) Polyphenolic antibacterials for food preservation:
review, challenges, and current applications. Foods 10:2469
20. Rodríguez-Daza MC, Pulido-Mateos EC, Lupien-Meilleur J, Guyonnet D, Desjardins Y, Roy
D (2021) Polyphenol-mediated gut microbiota modulation: toward prebiotics and further.
Front Nutr 8
21. Ray SK, Mukherjee S (2021) Evolving interplay between dietary polyphenols and gut micro-
biota—an emerging importance in healthcare. Front Nutr 8:1–18
22. Othman L, Sleiman A, Abdel-Massih RM (2019) Antimicrobial activity of polyphenols and
alkaloids in middle eastern plants. Front Microbiol 10
23. Bouarab-Chibane L, Forquet V, Lantéri P, Clément Y, Léonard-Akkari L, Oulahal N et al
(2019) Antibacterial properties of polyphenols: characterization and QSAR (quantitative
structure-activity relationship) models. Front Microbiol 10
24. Teodoro GR, Ellepola K, Seneviratne CJ, Koga-Ito CY (2015) Potential use of phenolic acids
as anti-Candida agents: a review. Front Microbiol 6:1–11
25. Delli Bovi AP, Marciano F, Mandato C, Siano MA, Savoia M, Vajro P (2021) Oxidative stress
in non-alcoholic fatty liver disease. An updated mini review. Front Med 8:1–14
26. Pérez-Jiménez J, Neveu V, Vos F, Scalbert A (2010) Identification of the 100 richest dietary
sources of polyphenols: an application of the phenol-explorer database. Eur J Clin Nutr
64:S112–S120
27. Tsao R (2010) Chemistry and biochemistry of dietary polyphenols. Nutrients 2:1231–1246
28. Bouarab Chibane L, Degraeve P, Ferhout H, Bouajila J, Oulahal N (2019) Plant antimicrobial
polyphenols as potential natural food preservatives. J Sci Food Agric 99
29. Stawarz-Janeczek M, Kryczyk-Poprawa A, Pytko-Polo J (2021) Antioxidant-rich natural raw
materials in the prevention and treatment of selected oral cavity and periodontal diseases.
Antioxidants 10:1–15
30. Makarewicz M, Drożdż I, Tarko T, Duda-Chodak A (2021) The interactions between polyphe-
nols and microorganisms, especially gut microbiota. Antioxidants 10:1–70
31. Romero C, Nardoia M, Arija I, Chamorro S (2021) Combining grape byproducts to maximise
biological activity of polyphenols in chickens. Animals 11:1–12
Dietary Natural Polyphenols Against Bacterial and Fungal Infections: An Emerging… 819
32. Cheng Y, Chen Y, Li J, Qu H, Zhao Y, Wen C et al (2019) Dietary β-sitosterol improves growth
performance, meat quality, antioxidant status, and mitochondrial biogenesis of breast muscle
in broilers. Animals 9
33. Taguri T, Tanaka T, Kouno I (2004) Antimicrobial activity of 10 different plant polyphenols
against bacteria causing food-borne disease. Biol Pharm Bull 27:1965–1969
34. de Silva OL, Garrett R, Monteiro MLG, Conte-Junior CA, Torres AG (2021) Pomegranate
(Punica granatum) peel fractions obtained by supercritical CO2 increase oxidative and colour
stability of bluefish (Pomatomus saltatrix) patties treated by UV-C irradiation. Food Chem
362:130159
35. Ekong MB, Iniodu CF (2021) Nutritional therapy can reduce the burden of depression man-
agement in low income countries: a review. IBRO Neurosci Rep 11:15–28
36. Kabra A, Sharma R, Hano C, Kabra R, Martins N, Singh BU (2019) Phytochemical composi-
tion, antioxidant, and antimicrobial attributes of different solvent extracts from myrica escu-
lenta buch.-ham. ex. d. Don leaves. Biomol Ther 9
37. Bracci L, Fabbri A, Del Cornò M, Conti L (2021) Dietary polyphenols: promising adjuvants
for colorectal cancer therapies. Cancers 13
38. Zhang W, Qi S, Xue X, Al Naggar Y, Wu L, Wang K (2021) Understanding the gastrointestinal
protective effects of polyphenols using foodomics-based approaches. Front Immunol 12:1–18
39. Drețcanu G, Iuhas CI, Diaconeasa Z (2021) The involvement of natural polyphenols in the
chemoprevention of cervical cancer. Int J Mol Sci 22
40. Tuli HS, Mittal S, Aggarwal D, Parashar G, Parashar NC, Upadhyay SK et al (2021) Path of
Silibinin from diet to medicine: a dietary polyphenolic flavonoid having potential anti-cancer
therapeutic significance. Semin Cancer Biol 73:196–218
41. Jantan I, Haque MA, Arshad L, Harikrishnan H, Septama AW, Mohamed-Hussein ZA (2021)
Dietary polyphenols suppress chronic inflammation by modulation of multiple inflammation-
associated cell signaling pathways. J Nutr Biochem 93:108634
42. Jiang H, Zhang W, Li X, Xu Y, Cao J, Jiang W (2021) The anti-obesogenic effects of dietary
berry fruits: a review. Food Res Int 147:110539
43. Knezevic S, Ghafoor A, Mehri S, Barazi A, Dziura M, Trant JF et al (2021) Catechin and other
catechol-containing secondary metabolites: bacterial biotransformation and regulation of car-
bohydrate metabolism. PharmaNutrition 17:100273
44. Marhuenda J, Silvia P, Victoria-montesinos D (2021) A randomized, double-blind, placebo-
controlled trial to determine the effectiveness of a polyphenolic extract (Hibiscus sabdariffa
and Lippia citriodora) for reducing blood pressure in prehypertensive and type 1 hypertensive
subjects. Molecules 26:1783
45. Płowuszyńska A, Gliszczyńska A (2021) Recent developments in therapeutic and nutraceuti-
cal applications of p-methoxycinnamic acid from plant origin. Molecules 26
46. Kowalska H, Kowalska J, Ignaczak A, Masiarz E, Domian E, Galus S et al (2021) Development
of a high-fibre multigrain bar technology with the addition of curly kale. Molecules 26:1–20
47. Augimeri G, Montalto FI, Giordano C, Barone I, Lanzino M, Catalano S et al (2021)
Nutraceuticals in the mediterranean diet: potential avenues for breast cancer treatment.
Nutrients 13
48. Vesely O, Baldovska S, Kolesarova A (2021) Enhancing bioavailability of nutraceutically used
resveratrol and other stilbenoids. Nutrients 13:1–15
49. Gomes MJC, Kolba N, Agarwal N, Kim D, Eshel A, Koren O et al (2021) Modifications in the
intestinal functionality, morphology and microbiome following intra-amniotic administration
(Gallus gallus) of grape (vitis vinifera) stilbenes (resveratrol and pterostilbene). Nutrients 13
50. Yamaguchi K, Itakura M, Kitazawa R, Lim SY, Nagata K, Shibata T et al (2021) Oxidative
deamination of lysine residues by polyphenols generates an equilibrium of aldehyde and
2-piperidinol products. J Biol Chem 297:101035
51. Scuto M, Trovato Salinaro A, Caligiuri I, Ontario ML, Greco V, Sciuto N et al (2021)
Redox modulation of vitagenes via plant polyphenols and vitamin D: novel insights for
820 B. Patra et al.
Purnima Paliwal, Sunita Panchawat, Rohini Trivedi and Devshree Gayakwad contributed equally
with all other contributors.
In epilepsy, patient observes a behavioral change that lasts only for a short period
that may be due to signs or symptoms (such as cognizance loss, stiffening, jerking),
caused by concurrent activity of neuron in the brain. Seizure arousal can be local-
ized (which affects one hemisphere of the brain), generalized (which affects both
the hemispheres) and unrevealed arousal [3]. Despite the fact that the cause of epi-
lepsy in many patients is unknown, seizures can be the outcome of about any insult
that unsettles the brain function. It can be caused by both genetic factor (Idiopathic)
and acquired factor (Symptomatic) [4]. These insults comprise acquired causes;
some may be peripheral to injury/surgery on the head, brain (intracranial tumor),
tuberculoma, cysticercosis, cerebral ischemia, etc. Epilepsy was divided into three
types by the International League Against Epilepsy (ILAE) in year 1989: Idiopathic
in which recurring seizures without detectable abnormalities, Cryptogenic epilepsy
with cause, and Symptomatic epilepsies had a recognizable cause [5]. Idiopathic
epilepsies constitute 40% of the epilepsies worldwide [6]. Idiopathic epilepsies are
further divided into idiopathic generalized and partial epilepsy. Some idiopathic
epilepsies occur due to inheritance by mutations in genes that code for iron chan-
nels. In symptomatic epilepsies, there are some genetic disorders that are responsi-
ble for neuropathological and neurocutaneous diseases or changes at the molecular
level: such as Angelman syndrome and Rett syndrome [7].
Thus, epilepsies have been classified variously; major types are described
in Fig. 1.
The main aim of antiepileptic drug therapy is to improve quality of life by control-
ling seizures and diminishing drug toxicity. It has been observed that about two-
thirds of epilepsy patients are cured with available antiepileptic drugs. Currently,
various first- and second-generation AEDs are in use as mentioned in Table 1, each
with its own limitations including side effects, tolerance, dependency, and extended
period toxicity [8]. The alternative path for the management of epilepsy can be by
the use of herbal sources and their active constituent having fewer side effects [9].
3 Worldwide Condition
Despite the incessant development of antiepileptic drugs, there are until now more
than 30% of patients accompanied by drug-resistant epilepsy, which leads to unusual
growth in the occurrence of disease and impermanence of epilepsy. Mechanism of
drug-resistant epilepsy is overexpression of efflux transporter, e.g., P-glycoprotein,
and altered expression of voltage-gated ion channels [11]. The most usual category
of drug-resistant epilepsy is temporal lobe epilepsy, generally cured by surgical
operation. Surgical treatment technique intended for mesial temporal lobe epilepsy
is intricate, and in surgeries like this, the patient’s well-being is a menace.Recently,
new drug targets emerge moderately, inclusive of glutamate neurotransmitters and
Ca2+ channels [12–15], transient receptor potential vanilloid-type 1 (TRPV1), Na+
K+ 2Cl− co-transporter (NKCC), etc. [16]. Sen et al. also established the increased
expression of NKCC1 in medically resistant refractory human epilepsy with hip-
pocampal sclerosis and focal cortical dysplasia [17]. Due to side effects possessed
by currently available AEDs, herbal remedies play a divine role in epilepsy
824 J. Pradhan et al.
Table 1 (continued)
Generation
of AEDs AED Efficacy spectrum Comments
Rufinamide Focal seizures, and Can cause loss of coordination and
drop attacks difficulty walking
associated with
LGS
Felbamate Focal seizures, and May cause a serious blood condition
drop attack or called aplastic anemia
assault associated
with LGS
Elisacarbazepine Focal seizures May precipitate or aggravate absence
acetate seizures and myoclonic seizures
Lacosamide Focal seizures May cause cardiac side effects
Pregabalin Focal seizures Can precipitate or aggravate myoclonic
seizures
Gabapentin Focal seizures Can precipitate or aggravate myoclonic
seizures
Tiagabine Focal seizures Can precipitate or aggravate absence
seizures and myoclonic seizures
Brivaracetam Focal seizures May cause clinically apparent drug
induced liver disease
Everolimus Seizures relate to Efficacy best documented against focal
tuberous sclerosis seizures associated with tuberous
complex only sclerosis complex
Stiripentol Seizures Only indicated for use in combination
associated with with clobazam and valproic acid against
Dravet syndrome tonic–clonic seizures associated with
only Dravet syndrome
The long-term use of currently available first- and second-generation AEDs is lim-
ited due to their adverse effects, withdrawal symptoms, and deleterious interactions
with other drugs. Furthermore, some of the available antiepileptic drugs may even
potentiate certain type of seizures. Given the wide availability of effective agents,
the toxicity and pharmacokinetic profile of an AED have become major factors in
the selection process. The following section defines various problems associated
with the use of existing AEDs:
826 J. Pradhan et al.
It has been divulged that women with epilepsy at the time of pregnancy with prena-
tal antiepileptic exposure have adverse effects in children. There are innumerable in
vitro evaluations that showed that antiepileptic drugs have an impact on neuronal
development [19]. Prenatal exposure to antiepileptic drugs with AEDs such as car-
bamazepine and valproate was found to exhibit adverse effects on verbal abilities.
During pregnancy or before birth, antiepileptics may produce growth-related com-
plications to the child. These types of developmental problems can be diagnosed by
the plasma nerve growth factor (NGF) level, which is the potential biomarker.
Periconceptional folate use may be important as a protective measure in at-risk
pregnancies [20].
Liver enzymes can entertain as markers of hepatocellular injury, e.g., aspartate ami-
notransferase (AST), alkaline phosphatase (ALP), alanine aminotransferase (ALT),
and gamma-glutamyl transferase (GGT). However, those enzymes are exaggerated
in liver disease, the exaggeration also can be subordinate to enzyme induction unac-
companied by hepatic pathology [26]. In certain, antiepileptic drugs have numerous
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 827
serious responses, as detected with carbamazepine (CBZ), valproic acid (VPA), and
phenytoin (PHT). However, to some extent relatively rare, when collated with dif-
ferent consistently known hepatotoxic drugs, the hepatotoxicity instigated by the
antiepileptic drug can cause death or an acute liver failure, which could critically
need liver transplantation. The hepatotoxicity instigated by antiepileptic drug hap-
pens either because of the production of reactive toxic metabolite/s or because of
induction of immune allergic reactions. Hepatotoxicity is analog with distinct clini-
cal indications of drug allergy (fever, rash, and eosinophilia). This reaction is char-
acteristic of CBZ and PHT. Additional idiosyncratic hepatotoxic reaction originates
from hepatotoxic metabolites because of abnormal metabolism. This reaction has
been observed with VPA [27].
About 50% of patients with epilepsy who take AEDs are observed to have bone
deformities. Manifestation of patients with AED-related bone diseases comprises
bone pain, muscle weakness, and fractures, with little or no torment. These manifes-
tations do not appear until the first fracture occurs [28]. For measuring the levels of
bone mineral density (BMD), there is a technique that is known as Dual-energy
X-ray absorptiometry (DXA) [29]. Abnormal BMD values were observed in one-
third to two-thirds of patients with epilepsy. However, it is understandable that
AEDs have an effect on bone metabolism and increased fractures are not clear, the
drug metabolism might play a dominant part in evolution of these adverse influ-
ences. Temazepam, a metabolite of diazepam via CYP3A4, was detected to be
increasing the possibility of fractures. Moreover, long-term use of CBZ may aug-
ment the danger of fracture and bone loss, instigate a status of reduction in bone and
mineral metabolism, increment in bone turnover, and diminution BMD. Continuing
use of VPA may upsurge the risks of bone damage [22]. TPM is also a carbonic
anhydrase inhibitor that might impede PTH-induced bone resorption, subsequent in
hypocalcemia. Juvenile rats treated with VB were observed to have reduced body
mass gain and repressed compact bone growth.
5.6.1 Cognitive Effects
5.6.2 Psychiatric Effects
Depression and anxiety are common among people with epilepsy [32]. The behav-
ioral adverse impact can occur with levetiracetam and also with topiramate,
zonisamide, and phenobarbital. Irritability and aggression are the topmost appre-
hensions with levetiracetam, often seen shortly after subsequent initiation. Psychosis
is sporadic. It may be preexisting, peri-ictal, or could be due to enforced stabiliza-
tion of brain action. Mood disorders, particularly anxiety and depression, are com-
munal comorbidities in epilepsy that may or may not be accompanied by AED
therapy. Patients should be evaluated for mood disorder earlier and often throughout
treatment as chronic dysthymia and depression disturb reporting of adverse effects
and seizure severity, also affecting the complete quality of life [33].
Many AEDs have limited use because of the risk for rare but serious effects that may
result in death. The use of felbamate and vigabatrin is limited because of the risk for
aplastic anemia and visual field deficits, respectively [34]. However, phenytoin, the
most widely used AED in the USA, may also cause hepatic failure.
5.8 Hypersensitivity
Maculopapular outbreak on the chest, inside elbow, and knee parts results in hyper-
sensitivity, which shows that the drug must be stopped as soon as possible. The rash
may lead to hypersensitivity, which can be lethal (e.g., Stevens–Johnson syndrome),
830 J. Pradhan et al.
needing hospitalization [33–36]. Lamotrigine rashes are high starting dose and
rapid escalation, as well as young age and concurrent valproate [31]. The drug
should be discontinued if the patient develops a rash. In the main surveying study of
1890 outpatients with epilepsy, researchers evaluated the rates of rash related to 15
AEDs [37]. Primary evaluating non-AED analysts of rash, they equated rash rates
for separate AEDs. The maximum rash rates arose with phenytoin (5.9%), lamotrig-
ine, (4.8%), and carbamazepine (3.7%). The lowermost rash rates arisen with felb-
amate, primidone, topiramate (all<1%), levetiracetam (0.6%), gabapentin (0.3%),
and valproate (0.7%).
5.9 Paediatric Issues
The probable for hostile events in children is superior than in grownups as young
children have immature detoxification mechanisms and a superior inconsistency in
dosing owed to a broader range of body size and weight. Valproate has been con-
cerned with hepatic toxicity in children, though phenobarbital, phenytoin, carbam-
azepine, oxcarbazepine, and lamotrigine cause advanced occurrences of rash in
children than adults [38, 39]. Metabolic acidosis, Hyperthermia, Nephrolithiasis,
and oligohydrosis are several disorders caused by Topiramate and Zonisamide in
children and adults [38–41]. Though, noteworthy metabolic acidosis in children can
lead to growth retardation [42] Gingival hyperplasia, an adverse event seen in
patients of all ages who take phenytoin, arises much more frequently in develop-
mentally impaired patients and young children [43].
Herbal medications are currently the greatest typical alternative to the allopathic
system of medication, which play a significant role in the treatment to control epi-
leptic seizures or complications caused by antiepileptic drugs. Nature is a wealthy
source of organic and chemical diversity. For decades, individuals with epilepsy are
using a diversity of botanicals and herbs, traditional schemes of medicine are preva-
lent in growing international countries, and up to 80% of the population depends on
traditional medicines for their prime healthcare requirements. Numerous plants
used for the remedy of epilepsy in exceptional structures of conventional medica-
tion have shown activity when tested on a contemporary day for the detection of
anticonvulsant activity. Herbal medicine is an area of complementary alternative
medicine that is readily amenable to empirical research [44, 45]. Numerous natural
drugs have impact on the central nervous system and are found to have antiepileptic
potential; some important ones, especially those studied during the last three
decades, are presented here along with their most important biologically active moi-
ety as reported in the literature (Table 3). The table also contains information about
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 831
7. Docosane (4)
8. N, N-Dimethyl valeramide O OH
9. 2,6,10,15-Tetramethyl heptadecane OH
HO O
O
(5)
O
(6)
O
(7)
H3C
CH3
(8)
N
O
(9)
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
OH
O
2 Alchornea Laxiflora 10. Quercetin-7,4′-disulphate O PTZ Whole plant
S
O OH
O O O
(Euphorbiaceae) 11. Quercetin O
S MES
OH
OH (10)
12. Quercetin-3′,4′-disulphate STR
OH O
13. Quercetin-3,4′-diacetate OH PIS
OH
14. Rutin HO O
NMDA
15. Quercetrin OH (11)
OH O
OH
O
O
S
OH
HO O O
(12)
O
OH O O S O
OH
OH
OCOCH3
HO O (13)
OCOCH3
OH O
OH
HO OH
OH O
HO
O
HO O OH
O
O OH
HO
OH (14)
OH
HO
O
HO O OH
O
O OH
HO
OH (15)
(continued)
833
Table 3 (continued)
834
HO
H
HO HO O
H
HO HO O
O O H
HO O O
H
OH O O
HO (19)
OH
HO
OH
HO OH
O O OH
O OH
(20)
O
OH
OH
OH
HO O
(21)
OH
OH O
HO
O
OH
HO
O
(22)
HO
OH
O
OH
HO
O (23)
J. Pradhan et al.
O
O O
HO (24)
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
5 Berberine 25. Berberine O N+ PTZ Berberine
O
(isoquinoline alkaloid) O
MES Hydrochloride solution
(25) O
KA
O
6 Carissa endulis 26. 3-O-acetyl chlorogenic acid OH
HO
(Apocynaceae) 27. Kaempferol 3-O-b-D glucopyranoside O OH
28. Quercetin 3-O-b-D glucopyranoside H3COCO OH O OH
29. Rhamnetin 3-O-b-D glucopyranoside (26)
R1
30. Isorhamnetin 3-O-b-D glucopyranoside
OH
31. Isorhamnetin 3-O-b-D
R1 O
glucopyranoside-(2″-1″)-rhamnopyranoside
32. Caredulis, 1- [1- [2-(2 hydroxypropoxy) OR3
propoxy] propane-2-yloxy] propane-2-ol OH O
33. (+)-butyl-α-L-rhamnoside
R1 R2 R3
27 H OH Glu
28 OH OH Glu
29 OH OMe Glu
30 OMe OH Glu
31 OMe OH Glu (2-1) Rha
OH
O O
O O OH
(32)
O O
HO H
OH
HO H
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
(33)
(continued)
835
Table 3 (continued)
836
O
O
(36) O
OH
O O O
(37)
OH
O
8 Cotyledon orbiculate 38. Cotyledoside C PTZ Whole plant
O
(Crassulaceae) 39. Orbicusides A O
PIC
O OH
40. Orbicusides B O BIC
O
O
41. Orbicusides C (38) NMDA
O
H
R2
OH
O O R1 O
O
O
R1 R2
39. O H
40. OH H
41. H OH
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
O
9 Cymbopogon 42. Citronellal (42) PTZ Essential oil from
winterians 43. Citronellol HO PIC leaves
(43)
44. Geraniol (44)
STR
HO
O O O
HO
O
O N
O O
(46)
O
HO
(47)
(continued)
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
837
Table 3 (continued)
838
HO
N
OO
(51)
O N
O
O
(52)
O
N O
O
(53)
OH
O
N O
(54) O
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
12 Ferula gummosa 55. Pinene MES Seed acetone extract
(Apiaceae) 56. α -thujene (55) PTZ
(56)
(58) OHOH
HO
O
OH
HO
O OH
(59)
HO
OH
O
OH
HO
(60)
O
(continued)
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
839
Table 3 (continued)
840
(63)
(64)
(65)
O
(68)
OH
(69)
O OH
(70)
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
HO
17 Leonotis leonurus 71. Apigenin PTZ Whole plant aqueous
O
(Lamiaceae) 72. Apigenin-8-C-β-glucoside OH BIC extract
HO (71)
73. Leonurine O PIC
OH NMDA
OH
HO OH
O
O OH
(72)
HO OH
O
O
HO
O N
O NH2
(73)
H2N
HO
18 Magnolia grandiflora 74. Magnolol MES Ethyl ether and
(Magnoliaceae) PTZ Hydroalcoholic extract
OH (74) of seed; bark
HO O OH
(79)
O (80)
N
HO NH2
O (81)
O
N
HO OH
O
21 Nardostachys 82. Nardin H PTZ Ethanol extract of roots
OH
jatamansi 83. Pyranocoumarine MES
H (82)
(Valerianaceae) H
O O O
(83)
O
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
O O O
22 Piliostigma 84. Piliostigmol PTZ Whole plant decoction
HO OH
reticulatum 85. 6,8-di-C-methylquercetin-3,30,7-trimethyl OH O STR
(84)
(Caesalpiniaceae) ether R1 NMDA
86. 6,8-di-C-methylquercetin-30-dimethyl ether O O O
R2
87. 30,6,8, -tri-C-methylquercetin-3,7-dimethyl OH OH
OH O R3
ether (85)
O
(94) O
O
(95)
(continued)
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
843
Table 3 (continued)
844
NH2
25 Sutherlandia 100. L-canavanine H2N N OH
PTZ Shoot aqueous extract
O
frutescens (Fabaceae) 101. Pinitol NH2 O
PIC
102. GABA (100) BIC
HO OH
103. Asparagines
O OH
HO OH
(101)
OH
O NH2
(102)
O
NH2
HO
NH2 O (103)
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
26 Taxus wallichiana 104. (E)-2-octen-1-ol OH PTZ Methanol leaf extract
(104)
105. n-pentacosane
106. Caryophyllene oxide
107. 1-octanol
108. Hexanoic acid
109. (Z)-3-hexenol (105)
O
(106)
OH
(107)
O
OH
(108)
OH
(109)
(continued)
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
845
Table 3 (continued)
846
O
HO
N N
HN N
O
O
O O
(112)
O H
O N
O N (113)
O
N O
O
N
O HO
N
N
H
(114)
J. Pradhan et al.
Name of the plant and Model used for
S. no family Active constituent Chemical structure evaluation Part of plant used
- - - N - -
O N
O H (115)
N
NH (116)
O O
HO O
O
O
O (119)
O
O
OH
a
Abbreviations: PTZ Pentylenetetrazole, MES Maximal Electroshock, NMDA N-Methyl-D-Aspartate, INH Isonicotinic acid, PIC Picrotoxin, STR Strychnine,
LPHE Lithium-Pilocarpine Hypoxic Episode, KA Kainic Acid, BIC Bicuculine, NKTM Nikethamide, TLE Temporal Lobe Epilepsy, SE Status Epilepticus, IPI
Initial precipitating injury, AEDs Antiepileptic drugs, HLTE Hind limb tonic extension, PHT Phenytoin, PB Phenobarbitone, GABA Gamma Amino Butyric Acid
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs
847
848 J. Pradhan et al.
Voltage-gated calcium channels (Fig. 3b) mediate calcium influx that both controls
neuronal excitability and regulates calcium-sensitive intracellular signaling path-
ways. From their closed/resting state, calcium channels open when the membrane
potential depolarizes to a threshold point, at which the inner voltage sensor moves
and the channel confirmation modified to an open-pore calcium-conducting state. In
these channels, ion conduction only takes place in open state. Voltage-gated calcium
channels are responsible for burst firing and rhythm generation. Their contribution
to neurons membrane potential is also noted. Their pharmacological classification is
as follows: High voltage-activated (HVA); L-type, N- type, R-type (responsible for
calcium entry and expressed in nerve terminals); P/Q, and low voltage-activated
(LVA); T-type (having voltage threshold, which is low, transient current generation,
burst firing pattern, and critical to pacemaker activity) [49]. The α1 subunits with
auxiliary subunits, which include β subunits (β1–β4), γ subunits (γ1–γ8), and α2–δ
subunits [50–52].
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 849
Voltage-Gated K + Channels
For limiting excitability in neural cells, K + channels (Fig. 3c) are the main targets.
α-Subunit, which comprises the ion-conduction pore, selectivity filter, and gating
apparatus, and various auxiliary subunits that serve modulatory roles, is responsible
for the formation of voltage-gated potassium channel. The typical K+ channel is a
tetramer of individual subunits, each one of which is homologous to a domain of the
pore-forming α or α1 subunits of voltage-gated Na+ or Ca2+ channels. There is clear
evidence that voltage-gated K+ channels are valid molecular targets for AEDs.
850 J. Pradhan et al.
HCN Channels
The mammalian ClC gene family encodes nine Cl channels with diverse functions
in plasma membranes and intracellular organelles. One of these channels, ClC-2, a
homodimeric channel found in neurons and glia, has been implicated in epilepsy
and thus may be an important target for AEDs.
Fig. 4 Ligand-gated ion channel; a.: Membrane topology diagram of cys-loop receptors- GABA,
Ach, and Glycine receptor channels; b. Glutamate receptor channel; c. ATP-gated channels
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 851
GABAA Receptors
Gamma amino butyric acid (GABA) is the main inhibitory neurotransmitter in the
brain. Inhibitory neurons that make use of GABA as neurotransmitter are found
throughout the brain, but in any region they may comprise a wide range of morpho-
logical and functional types that participate in different circuits with principal neu-
rons. Chemical agents that impair GABAergic inhibition are powerful convulsants,
and therefore, an increase in activity of GABA is associated with anticonvulsant
action. GABAA receptors have a pentameric structure, in which around central Cl−
pore five subunits are arranged like spokes of a wheel. Nineteen distinct genes
encode 19 subunits (α1–6, β1–3, γ1–3, δ, ε, θ, π, ρ1–2). Each subunit has four trans-
membrane segments, with both the amino and carboxy termini located extracellu-
larly. These extracellular segments form the recognition sites (two per channel) for
GABA and also, in some channel types, the recognition site (one per channel) for
benzodiazepine-like allosteric modulators [54]. The emerging understanding of the
different roles of phasic and tonic inhibition in epileptic phenomena may suggest
improved approaches to targeting GABAA receptors for epilepsy therapy.
Glycine Receptors
Ionotropic glutamate receptors are tetrameric structures (Fig. 4b), which encompass
three receptor families identified by the agonists that selectively activate them:
AMPA, kainate, and NMDA. Each of the families has a specific set of subunits: four
for the AMPA receptors, five for the kainite receptors, and seven for the NMDA
receptors [57]. These channels mediate most of the fast excitatory transmission in
the central nervous system and are thus involved in all brain functions. Their exces-
sive activation plays a large role in epileptic phenomena, including the paroxysmal
depolarization shift and seizure discharges.
NMDA Receptors
NMDA receptors are blocked in a voltage-dependent fashion by means of Mg2+,
such that the open channels are in large part impermeable to ion flow at negative
membrane potentials (near the resting potential), due to retention of Mg2+ in the
channel pore.
AMPA Receptors
AMPA receptors are composed of four kinds of subunits, designated GluR1–4
(alternatively GluR A–D), which intrigate to form tetramers. AMPA receptors serve
as the primary mediators of fast excitatory neurotransmission in the mammalian
central nervous system, and changes in their cellular expression underlie forms of
synaptic plasticity. AMPA receptor antagonists may have antiepileptogenic activity,
if only by virtue of their capability to powerfully suppress the seizures.
Acid-sensing ion channels are a family of proton-gated cation channels related to the
epithelial Na+ channels, which have diverse functions in metazoan cells. Six isoforms
have been cloned; these are widely expressed as homomeric or heteromeric channels
in the central and peripheral nervous systems. ASICs in sensory neurons are believed
to be involved in nociception when injury or inflammation causes acidification. The
functional roles of ASICs in the brain are less well understood.
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 853
GPCRs comprise the largest set of therapeutic drug targets for known medicinal
agents. All GPCRs have an extracellular N-terminal, an intracellular C-terminal,
and seven α-helical transmembrane sections (Fig. 5). They are separated into three
chief classes, out of them of relevant importance here are the metabotropic gluta-
mate receptors (mGluRs) and the GABAB receptors, which play important roles in
controlling excitability at glutamatergic and GABAergic synapses. Any peptide
neurotransmitters, such as neuropeptide Y, galanin, somatostatin, and the enkepha-
lins and endorphins, which act at GPCRs, are inclusive in epileptic phenomena.
The metabotropic glutamate receptors are a family of eight G-protein receptors that
are categorized into three groups defined by their sequence homology, second mes-
senger effects, and common pharmacology. There is little evidence to indicate that
genetic alterations of mGluRs play a role in the pathogenesis of epilepsy.
GABAB Receptors
7.2.1 GABA-Transaminase
GABA released from synapse is transported into astrocytes and nerve terminals,
where it is either taken up into synaptic vesicles or metabolized by two mitochon-
drial enzymes, GABA-transaminase (4-aminobutyrate-2-oxoglutarate aminotrans-
ferase) and succinic semialdehyde dehydrogenase, to yield succinate, a tricarboxylic
acid cycle intermediate. It was shown that inhibitors of GABA-transaminase raise
brain GABA content and inhibit seizures in rodents. But at the same time, it is also
reported that chronic inhibition of GABA-transaminase has a risk of retinal damage,
and that is why the utility of GABA-transaminase as an AED target remains
uncertain.
7.2.2 Carbonic Anhydrase
Computer-aided drug design (CADD) approaches are useful tools to interpret and
guide experiments to expedite the drug design process. CADD is of two types, viz.
structure-based Drug Design (SBDD) and ligand-based Drug Design (LBDD). In
structure-based, the structural knowledge of target is considered, and in ligand-
based knowledge of known ligands with their bioactivity is considered. There are
different methods available for identifying new drugs, which include 3D-QSAR,
Docking studies, MD simulation studies, and ADME properties [62]. Figure 6
shows the CADD application for searching and investigating new drugs.
Fig. 6 Application of computer-aided drug design (CADD) for the design of new drug
856 J. Pradhan et al.
In cases where the 3D structure of the target protein is lacking, information taken
from a set of ligands active against a target (receptor or enzyme) can be used to
detect important structural and physicochemical properties (molecular descriptors)
responsible for the detected biological activity. Here, there is an assumption that
structurally similar compounds display similar biological responses and interaction
with the target. The compound set should encompass a wide range of concentrations
(at least four orders of magnitude) to generate a reliable ligand-based screening
model. Common ligand-based design techniques are:
(a) Quantitative structure–activity relationship (QSAR)
(b) Pharmacophore modeling
(c) Compound selection
(d) Specificity
Docking-Based Virtual Screening Molecular docking is used to predict the binding
mode of ligands within the binding site of target proteins. Various docking software
are available inclusive of Schrodinger, Autodock Vina, Molegro, etc., which differ
based on scoring function and conformational sampling. Docking calculation is
done to investigate the binding affinity between compounds and targets. Different
target proteins are downloaded from the Protein Data Bank.
References
9. Giridhar BN, Srinivasu CC, Raju PT, Lakshmana DN (2015) Estimation of trace elements
concentration in anti-epileptic medicinal plants by using EDXRF. Int J Sci Res (IJSR)
4(2):211–216. https://doi.org/10.21275/SUB15931
10. Beghi E (2019) The epidemiology of epilepsy. Neuroepidemiology:1–7. https://doi.
org/10.1159/000503831
11. Patrick K, Steven SC, Martin BJ (2011) Drug resistance epilepsy. Eng J Med:919–926. https://
doi.org/10.1056/NEJMra1004418
12. Rajakulendran S, Hanna MG (2016) The role of calcium channels in epilepsy. Cold Spring
Harb Perspect Med. https://doi.org/10.1101/cshperspect.a022723
13. Kohl M, Paulsen O (2010) The roles of GABAB receptor in cortical network activity. Adv
Pharmacol:205–229. https://doi.org/10.1016/S1054-3589(10)58009-8
14. Go T (2004) Carbamazepine-induced IgG1 and IgG2 deficiency associated with B cell matura-
tion defect. Seizure:187–190. https://doi.org/10.1016/S1059-1311(03)00086-4
15. Lasoń W, Chlebicka M, Rejdak K (2013) Research advances in basic mechanisms of sei-
zures and antiepileptic drug action. Pharmacol Rep:787–801. https://doi.org/10.1016/
S1734-1140(13)71060-0
16. Hass M, Forbush B III (2009) The Na-K-Cl cotrnasporte of secretory epithelia. Annu Rev
Physiol 62:15–34
17. Sen A, Martinian L, Nikolic M, Walker MC, Thom M, Sisodiya SM (2007) Increased NKCC1
expression in refractory human epilepsy. Epilepsy Res:220–227. https://doi.org/10.1016/j.
eplepsyres.2007.01.004
18. Arun SK, Ekta R, Abdul W, Satyendra RK (2015) Pharmacoresistant epilepsy: a current update
on non-conventional pharmacological and non-pharmacological interventions. J Epilepsy Res
5(1). https://doi.org/10.14581/jer.15001
19. Meador KJ, Gilliam FG, Kanner AM, Pellock JM (2001) Cognitive and behavioral effects
of antiepileptic drugs. Epilepsy Behav 2:SS1–SS17. https://doi.org/10.1006/ebeh.2001.0235
20. Karaya A, Burak O, Selçuk İ, Co M (2019) Epilepsy & behavior serum NGF levels may be
associated with intrauterine antiepileptic exposure-related developmental problems. Epilepsy
Behav:60–66. https://doi.org/10.1016/j.yebeh.2019.05.013
21. Dana-Haeri J, Oxley J, Richens A (1984) Pitutary responsiveness to gonadotrophin-releasing
and thyrotrophin-releasing hormone in epileptic patients reciving carbamazepine or phenytoin.
Clin Endocrinol:163–168. https://doi.org/10.1111/j.1365-2265.1984.tb00071.x
22. Spina E, Perucca E (2002) Clinical significance of pharmacokinetic interactions between anti-
epileptic and psychotropic drugs. Epilepsia:37–44. https://doi.org/10.1046/j.1528-1157.2002
.043s2037.x
23. Vecht JC, Wagner LG, Wilms BE (2003) Interaction between antiepilectic and chemotherapeu-
tic drugs. Lancet Neurol:404–409. https://doi.org/10.1016/S1474-4422(03)00435-6
24. Isojärvi JIT (1993) Serum lipid levels during carbamazepine medication. Arch Neurol:17–20.
https://doi.org/10.1001/archneur.1993.00540060030012
25. Mintzer S, Mattson RT (2009) Should enzyme-inducing antiepileptic drugs be considered
first-line agents. Epilepsia:42–50. https://doi.org/10.1111/j.1528-1167.2009.02235.x
26. Ahmed SN, Siddiqi AZ (2006) Antiepileptic drugs and liver disease. Seizure:156–164. https://
doi.org/10.1016/j.seizure.2005.12.009
27. Arroyo S, Morena A (2001) Life-threatening adverse events of antiepileptic drugs. Epilepsy
Res:155–174. https://doi.org/10.1016/S09201211(01)00306-0
28. Sheth RD (2004) Bone health in epilepsy. Lancet Neurol:516. https://doi.org/10.1016/
S1474-4422(04)00848-8
29. Musa A, Haluk S, Ercan M (2003) Evaluation of bone mineral density with dual x-ray absorp-
tiometry for osteoporosis in children with bladder augmentation. J Pediatr Surg:230–232.
https://doi.org/10.1053/jpsu.2003.50050
30. Ludwig B, Marsan CA (1975) Clinical ictal pattern in epileptic patients with occipital electro-
encephalographic foci. Neurology:463–471. https://doi.org/10.1212/wnl.25.5.463
860 J. Pradhan et al.
31. Hermann B, Meador JK, Gaillard DW, Cramer AJ (2010) Cognition across the lifes-
pan: antiepileptic drugs, epilepsy, or both? Epilepsy Behav:1–5. https://doi.org/10.1016/j.
yebeh.2009.10.019
32. Blumer D, Montouris G, Hermann B (1995) Psychiatric morbidity in seizure patients on
a neurodiagnostic monitoring unit. Neuropsychiatry Clin Neurosci:445–456. https://doi.
org/10.1176/jnp.7.4.445
33. Cramer JA, Mintzer S, Wheless J, Mattson RH (2010) Adverse effects of antiepileptic drugs:
a brief overview of important issues. Expert Rev Neurother:885–891. https://doi.org/10.1586/
ern.10.71
34. Besag Frank MC (2004) Behavioural effects of the newer antiepileptic drugs: an update.
Expert Opin Drug Saf:1–8. https://doi.org/10.1517/14740338.3.1.1
35. Parker WA, Shearee CA (1979) Phenytoin hepatotoxicity: a case report and review.
Neurology:175–178. https://doi.org/10.1212/wnl.29.2.175
36. Anderson GD (2002) Children versus adults: pharmacokinetic and adverse-effect differences.
Epilepsia:53–59. https://doi.org/10.1046/j.15281157.43.s.3.5.x
37. Arif H, Buchsbaum R, Weintraub D, Koyfman S, Salas-Humara C, Bazil CW, Resor SR,
Hirsch LJ (2007) Comparision and predictors of rash associated with 15 antiepileptic drugs.
Neurology:1701–1709. https://doi.org/10.1212/01.wnl.0000261917.83337.db
38. Perucca P, Gilliam GF (2012) Adverse effects of antiepileptic drugs. Lancet Neurol:792–802.
https://doi.org/10.1016/S1474-4422(12)70153-9
39. Fricke-Galindo I, Jung-cook H, Lerena A, Lopez-Lopez M (2018) Pharmacogenetics of
adverse reactions to antiepileptic drugs. Neurologia:165–176. https://doi.org/10.1016/j.
nrl.2015.03.005
40. Moore TJ, Weiss SR, Kaplan S Blaisdell CJ (2002) Reported adverse drug events in infants
and children under 2 years of age. Pediatrics:1–5. https://doi.org/10.1542/peds.110.5.e53
41. Mathis LL, Lyasu S (2007) Safety monitoring of drugs granted exclusivity under the best phar-
maceuticals for children act: what the FDA has learned. Clin Pharm Therap:133–134. https://
doi.org/10.1038/sj.clpt.6100285
42. McSherry E (1978) Acidosis and growth in nonuremic renal disease. Kidney Int:349–354.
https://doi.org/10.1038/ki.1978.135
43. Camfield P, Camfield C (2006) Monitoring for adverse effects of antiepileptic drugs.
Epilepsia:31–34. https://doi.org/10.1111/j.15281167.2006.00657.x
44. Liu W, Ge T, Pan Z, Leng Y, Lv J, Li B (2017) The effects of herbal medicine on epilepsy.
Oncotarget:48385–48397. https://doi.org/10.18632/oncotarget.16801
45. Pradhan J, Sunita P (2018) Herbal therapies for epilepsy: chemistry, biology and potential
applications of selected plants and compounds. Chem Biol Interface 8(4):205–224
46. Tripathi M, Reddy P, Rawat D (2014) Chemistry and biology interface. Chem Biol 4:1–22
47. Ragsdale D, McPhee J, Scheuer T, Catterall W (1994) Molecular determinants of state-
dependent block of Na+ channels by local anesthetics. Science:1724–1728. https://doi.
org/10.1126/science.8085162
48. Ragsdale D, McPhee J, Scheuer T, Catterall W (1996) Common molecular determinants of
local anesthetic, antiarrhythmic, and anticonvulsant block of voltagegated Na+ channels. Proc
Natl Acad Sci:9270–9275. https://doi.org/10.1073/pnas.93.17.9270
49. Yarov-Yarovoy V, Brown J, Sharp EM, Clare JJ, Scheuer T, Catterall WA (2001) Molecular
determinants of voltage-dependent gating and binding of pore-blocking drugs in transmem-
brane segment IIIS6 of the Na+ channel alpha subunit. J Biol Chem:20–27. https://doi.
org/10.1074/jbc.M006992200
50. Drew CDM (2000) Instant pharmacology. Postgrad Med J 76(896):381–381a. https://doi.
org/10.1136/pmj.76.896.381a
51. Ffrench-Mullen JM, Barker JL, Rogawski MA (1993) Calcium current block by (−)-pen-
tobarbital, phenobarbital, and CHEB but not (+)-pentobarbital in acutely isolated hippo-
campal CA1 neurons: comparison with effects on GABA-activated Cl- current. J Neurosci
13(8):3211–3221. https://doi.org/10.1523/jneurosci.1308-03211.1993
Phyto-Constituents as Potential Leads for the Development of Novel Antiepileptic Drugs 861
52. Stefani A, Spadoni F, Siniscalchi A, Bernardi G (1996) Lamotrigine inhibits Ca2+ currents
in cortical neurons: functional implications. Eur J Pharmacol 307:113–116. https://doi.
org/10.1016/0014-2999(96)00265-8
53. Pisani A, Bonsi P, Martella G, De Persis C, Costa C, Pisani F, Bernardi G, Calabresi P (2004)
Intracellular calcium increase in epileptiform activity. Epilepsia 45(7):719–728. https://doi.
org/10.1111/j.0013-9580.2004.02204.x
54. Hainsworth HA, Nicolle MCL, Alexey P, Toni S, Andrew RD (2003) Actions of sipatrigine,
202W92 and lamotrigine on R-type and T-type Ca2+ channel currents. Eur J Pharmacol
467(1–3):77–80. https://doi.org/10.1016/S0014-2999(03)01625-X
55. Arias RL, Bowlby RM (2005) Pharmacological characterization of antiepileptic drugs and
experimental analgesics on low magnesium-induced hyperexcitability in rat hippocampal
slices. Brain Res 1047(2):233–244. https://doi.org/10.1016/j.brainres.2005.04.052
56. Monaghan EP, McAuley JW, Data JL (1999) Ganaxolone: a novel positive allosteric modu-
lator of the GABA(A) receptor complex for the treatment of epilepsy. Expert Opin Investig
Drugs 8(10):1663–1671. https://doi.org/10.1517/13543784.8.10.1663
57. Scheffer IE, Bhatia KP, Lopes-Cendes I, Fish DR, Marsden CD, Andermann E, Andermann
F, Desbiens R, Keene D, Cendes F, Manson JI, Constantinou JEC, Mclntosh A, Berkovic SF
(1995) Autosomal dominant nocturnal frontal lobe epilepsy. Brain 118(1):61–73. https://doi.
org/10.1093/brain/118.1.61
58. Steinlein OK, Mulley JC, Propping P, Wallace RH, Phillips HA, Sutherland GR, Scheffer
IE, Berkovic SF (1995) A missense mutation in the neuronal nicotinic acetylcholine receptor
α4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy. Nat Genet
11(2):201–203. https://doi.org/10.1038/ng1095-201
59. Mayer ML, Armstrong N (2004) Structure and function of glutamate receptor ion channels.
Annu Rev Physiol 66(1):161–181. https://doi.org/10.1146/annurev.physiol.66.050802.084104
60. Bowery NG, Smart TG (2006) GABA and glycine as neurotransmitters: a brief history. Br J
Pharmacol 147:109–119. https://doi.org/10.1038/sj.bjp.0706443
61. Bowery NG, Hill DR, Hudson AL, Middlemiss DN, Shaw J, Turnbull M (1980) (−)Baclofen
decreases neurotransmitter release in the mammalian CNS by an action at a novel GABA
receptor. Nature 283(5742):92–94. https://doi.org/10.1038/283092a0
62. Brogi S (2019) Computational approaches for drug discovery. Molecules 24(17):3061. https://
doi.org/10.3390/molecules24173061
Role of Polyphenols in Cardiovascular
Diseases
1 Introduction
Polyphenols are naturally occurring compounds found in a broad range of foods and
beverages. Polyphenol concentrations in fresh fruit range between 200 and 300 mg
per 100 g. They contain a lot of polyphenols, which may be found in the goods
made from these fruits. In a serving of red wine, tea, or coffee, polyphenols may
vary from 100 to 200 mg. Chocolate and cocoa beans contain high levels of poly-
phenols [1–3].
Polyphenols, plant secondary metabolites, have two primary functions: UV pro-
tection and disease defense. Color, flavor, aroma, and oxidative stability are all
directly influenced by polyphenols. For cancer, cardiovascular disease, diabetes,
osteoporosis, and neurological illnesses, epidemiological studies and meta-analyses
H. Chopra
Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
S. Bibi
Department of Biosciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
Y. K. Mohanta
Nano-biotechnology and Translational Knowledge Laboratory, Department of Applied
Biology, University of Science and Technology Meghalaya (USTM), Techno City, Baridua,
Ri-Bhoi, Meghalaya, India
e-mail: [email protected]
S. Kumari
Department of Applied Biology, University of Science and Technology Meghalaya,
Ri-Bhoi, India
A. A. Baig (*)
University Institute of Public Health, Faculty of Allied Health Sciences, The University of
Lahore, Lahore, Pakistan
Institute of Molecular Biology & Biotechnology, The University of Lahore, Lahore, Pakistan
suggest that dietary polyphenols may be beneficial [4–17]. Polyphenols and other
dietary phenolics are increasingly studied for their potential health benefits. Food
polyphenols and their effects on health and sickness are the focus of this research
project. Phytochemicals are substances that plants make for their own benefit. The
phrase “essential nutrient” may describe non-essential plant components. Individual
nutrients’ impacts on health remain unknown, but the benefits of eating a diet rich
in fruits and vegetables have long been recognized by the general public. If you eat
a plant-based diet, the health benefits of specific phytochemicals are linked to those
of the foods that contain those phytochemicals [5–7, 9].
Polyphenols are secondary compounds found in plants. Flavonoids (flavonols,
flavones, flavanones, isoflavones, proanthocyanidins) and stilbenes and lignans are
molecules that make up phenolic acids (hydroxybenzoic and hydroxycinnamic
acids). Phenolic chemicals increase the flavor and nutritional value of fruits in vari-
ous ways [18–22]. The antioxidant characteristics of phenolic compounds might
help protect against LDL, platelet aggregation, and damage to red blood cells [23].
Antimutagens, anticarcinogens, antimicrobials, and clarifiers are all phenolics’
other properties [24–33]. These chemicals impact the color, astringency, and bitter-
ness of red wine [34, 35]. Fruit and vine stems and yeast metabolism may both
produce these by-products. Phenolics also serve as oxygen reservoirs and browning
substrates. Polyphenols’ biological properties are influenced by their bioavailabil-
ity. It is governed by their chemical structure as to how quickly they are absorbed in
the intestines. Green tea’s catechins and citrus fruits’ flavanones have a high bio-
availability [36]. An assessment of intestinal absorption may be made by looking at
the antioxidant capacity of plasma following polyphenol-rich food consumption.
Flavonoids’ plasma concentration decreases after they are ingested. Because plasma
albumin has a significant affinity for quercetin, it has a longer elimination half-life.
Antioxidants such as glycosylated flavonoids are found in abundance [37].
Cardiovascular disease is influenced by both environmental and hereditary factors.
Variables including physical activity, smoking, and a diet heavy in saturated fat
increase the prevalence of cardiovascular disease (CVD) [38]. As a result, it is dif-
ficult to pinpoint a single reason for many ailments in this population. There is
strong evidence that diets high in polyphenols, such as those found in tea and cocoa,
may improve cardiovascular health [39]. The consumption of flavanols, flavonols,
and flavones was associated with an increased risk of coronary heart disease (CHD)
[40–43]. Flavanones and anthocyanins reduced cardiovascular disease deaths [44–
47]. Those who drank three cups of tea a day reduced their risk of cardiovascular
disease by 11% [48].
Many people argue over which polyphenols are best for reducing heart disease.
Flavonoids, which are found in chocolate and soy, have lower cardiovascular dis-
ease risk [39]. Reduced blood pressure, increased endothelial function, and
decreased platelet adhesion have been linked to antioxidant effects on low-density
lipoprotein (LDL). A diet high in flavanols from cocoa may lessen the risk of car-
diovascular disease and hypertension [39]. Hypertension risk may be reduced by
Role of Polyphenols in Cardiovascular Diseases 865
drinking one cup of black tea daily [49]. Short-term tea consumption had no effect
in lowering blood pressure [50].
Endothelial cell bioavailability of nitric oxide may be influenced by polypheno-
lic chemicals, which impact an enzyme known as nitric oxide synthase [51, 52].
Polyphenols may cause endothelium-dependent relaxation [9]. Black tea and purple
grape juice have been shown to reduce platelet activation and aggregation [53–55].
Flavanols and flavonoids may be used to prevent vascular damage [56]. Polyphenols
have been shown to positively impact cardiovascular health via a wide range of
epidemiological studies (as shown in Fig. 1) [57]. Overall mortality risk was
inversely linked to polyphenol intake from diverse sources, such as red wine,
almonds, and olive oil, in the PREDIMED trial [58, 59]. Polyphenol intake was a
substantial risk factor for heart disease in the same study. A lot of factors must be
considered while analyzing observational data. It is possible that eating more
polyphenol-rich plant foods and reducing your diet of animal foods will improve
your health. Polyphenol consumption estimates may be skewed by a recall, portion
size, and the likely polyphenol content of similar meals. In this kind of investiga-
tion, several variables might make it difficult to pinpoint the underlying reason. An
observational study is the first step in conducting controlled intervention research.
Clinical trials that include specific phenolic compounds and phenolic-rich extracts
or meals should be considered [58, 59].
2.1 Diabetes
with type 2 diabetes who had intensive or regular glycaemic therapy had no change
in heart failure rates, according to a meta-analysis that included 37,229 individuals
[65]. A cohort study of 271,174 people with T2DM from the Swedish National
Diabetes Register and 1,355,870 matched controls found that 22 people under the
age of 55 with T2DM who had several risk factors (HbA1c, LDL-cholesterol, blood
pressure, albuminuria, and smoking) outside the target had the highest risk of hos-
pitalization for HF.
Hospitalization for heart failure was associated with atrial fibrillation, a high
BMI, and higher HbA1c and renal function [66]. Diabetes-related high blood pres-
sure (HDH) may cause a decreased or increased ejection fraction (HEF) (HFpEF).
In individuals with type 2 diabetes, HFpEF has overtaken HFrEF as the most fre-
quent form of ventricular dysfunction, and T2DM is becoming an increasingly
important role in the pathogenesis of LV dysfunction [63, 67].
2.2 Cholesterol
As a result, men who begin smoking at a young age are more likely to get the
disease [72]. The vast majority of peripheral artery disease-caused amputations
result from diabetes and smoking. Atherosclerosis causes the walls of blood vessels
to become thinner over time. Symptoms will begin to appear within a few months.
Fontaine and Rutherford are utilized to determine the severity of a patient’s illness
[73]. Studies have shown that the ankle-brachial index (ABI) is a risk factor for
coronary artery disease in patients with lower limb illnesses. In contrast, an abnor-
mally low ABI indicates a significant medical problem.
According to the 2017 European Society of Cardiology (ESC) recommenda-
tions, those with an initial LDL level of 70–135 mg/dL should have their cholesterol
lowered to 70 mg/dL or reduce their LDL levels by 50% [74]. Statin treatment has
been found to reduce overall and cardiovascular mortality in individuals with
peripheral arterial disease. There was a strong correlation between abdominal aortic
aneurysms and the presence of atherosclerotic cardiovascular disease. Patients with
abdominal aneurysms were typically discovered to have additional cardiovascular
problems. The risk of developing an aortic aneurysm increases with increasing age,
smoking, and having a male significant other [75]. While intimal atherosclerosis
and thrombosis are frequent, elastin fragmentation and persistent inflammation only
occur in aortic aneurysms.
2.3 Smoking
The risk of coronary heart disease (CHD) related to cigarette smoking may be quan-
tified using RR and excess risk [76]. Nonsmokers are more likely to die from coro-
nary heart disease (CHD) than smokers, according to the risk ratio (RR). To explain
the difference in illness rates between smokers and nonsmokers, the phrase “extra
risk” is employed.
Depending on the risk estimate you choose, your chance of developing coronary
heart disease (CHD) increases or decreases over time. The Cancer Prevention Study
II (CPS-II) data display the RRs and excess mortality rates by age group [76]. As the
RRs became older, they began to decrease rapidly (35–54 years old). According to
these new studies, tobacco-related heart attacks seem to be less prevalent among the
elderly. On the other hand, smokers’ mortality rates from coronary heart disease
(CHD) have been demonstrated to be inconsistent. People under the age of 35 are at
increased risk of coronary heart disease due to a lower mortality rate from CHD and
a higher incidence of coronary events among smokers. There was an increase in
smoking-related mortality from coronary heart disease (CHD) despite the RR
decreasing as people aged.
Role of Polyphenols in Cardiovascular Diseases 869
3 Phenolics in CVD
denying that isorhamnetin was the most intriguing molecular entity discovered.
Doxorubicin’s antitumor efficacy was boosted, as well as its ability to protect
against cardiotoxicity [114, 115]. Centaurea borysthenica Gruner and Centaurea
daghestanica (Lipsky) Wagenitz have recently been studied for their phenolic con-
tent and cardioprotective effects [116]. Doxorubicin-exposed rat cardiomyocytes
were shown to be protected by both C. borysthenica, and C. daghestanica extracts.
In this investigation, C. daghestanica methanolic extracts had no effect on the effec-
tiveness of doxorubicin.
In addition, Alhaider’s team researched the cardioprotective properties of the
date palm, known in Arabic as Nakl (date). The total flavonoid, total phenolic, and
antioxidant capabilities of four date palm fruit extracts from eastern Saudi Arabia
were investigated using in vivo mice myocardial infarction models. Antioxidants
and heart-protective flavonoids found in fruit extracts have been linked to tissue
recovery after ischemia by mobilizing circulating progenitor cells from the bone
marrow to the site of myocardial infraction [117]. H9c2 cardiac cell lines were
exposed to oxidative stress, LDL oxidation, HMG-CoA reductase, angiotensin-
converting enzyme regulation in vitro, and extract fractions of Syzygium cumini (L.)
Skeels seeds showed cardioprotective effects. Acids such as ellagic, cinnamic, feru-
lic, and quercetin make up the group of compounds. In experiments on H9c2 cardio-
myocytes, these fractions reduced oxidative stress, and molecular docking showed
a favorable link between phytochemical substances and enzymes essential for
avoiding cardiovascular disorders [118]. The heart-protective effects of Marrubium
vulgare L., an Iranian medicinal herb, were investigated in an in vivo Wistar rat
model. To determine the aqueous fraction of extract’s total phenolic and flavonoid
content, Langendroff analysis was utilised [119]. These two primary chemicals
from Aspalathus linearis (Burm.f.) R. Dahlgren may prevent myocardial infarction
caused by chronic hyperglycemia [120]. By opening the Ca2+-activated K+ channel
and activating protein kinase C, the isoflavone puerarin protects the heart against
damage caused by ischemia and reperfusion. In an in vivo rat model with cardiovas-
cular risk factors, Tian and his colleagues found that polyphenolic extracts from
apple peels were more cardioprotective than extracts from apple meat [121]. The
skin of an apple has more total flavonoids and total phenolics.
Human and animal studies have linked moderate alcohol use, especially red
wine, to an elevated risk of cardiovascular disease (CVD). Compared to those who
never drank at all, moderate alcohol intake may reduce the risk of death from heart
disease and other causes by 6–5 drinks per day in case–control studies and five
drinks per day in cohort studies [122]. It contains 14 grams of alcohol per serve.
Moderate use of alcohol offers more favorable effects than abstinence, but these
advantages are lost when consumption is excessive. Women benefit from a daily
alcohol intake of 3–30 grams, while males benefit from daily alcohol consumption
of 12–60 grams [123]. Rimm et al. found that drinking between 30 and 50 g of
alcohol per day may reduce the chance of having ischemic heart disease by 42%
[124]. Women in their 40 s and 50 s who consumed 5–24 g of alcohol per day low-
ered their risk for ischemic heart disease by 40% and total stroke by 40–50%,
872 H. Chopra et al.
4 Flavonoids
Egg yolk, red meat, and fish high in choline and betaine produce trimethylamine
N-oxide (TMAO). Proximal cecum and cecum anaerobic bacteria digest dietary
fibers with a bit of aid from the breakdown of protein and peptides to create them
[137]. Insulin sensitivity, glucose and fat metabolism are all affected by the pres-
ence of short-chain fatty acids (SCFAs). It has been established that SCFAs may
lower cardiovascular and metabolic disease risk factors [138]. TMAO and SCFA,
two bioactive chemicals generated by genetically modified organisms (GMOs), are
linked to chronic cardiovascular disease (CVD) [139, 140]. Thrombosis, hyperten-
sion, and heart failure have all been linked to alterations in the microbiota in the
human gut (as shown in Fig. 2) [142]. It has recently been demonstrated that GPCRs
may regulate blood pressure through activating GPRs, such as GPR3 and Olr78
(Olfr78). It is SCFAs that energize GPR41, GPR43, and Olfr78. GPR41 is an
Table 1 Natural chemical compounds studied against cardiovascular diseases worldwide
Stilbene
Trimethylamine oxide
resveratrol
Quercetin
876 H. Chopra et al.
Fig. 2 Cardiovascular disease and flavonoid metabolism are linked to gut bacteria. Ingesting cho-
line from dietary sources (such as cholesterol, phospholipids, and l-carnitine) allows the GM to
create TMA. Flavin monooxygenase is oxidized to generate TMAO in the liver, which subse-
quently enters the systemic circulation through the hepatic circulation. Foam cells and platelets
build when TMAO levels rise, disrupting cholesterol transport and encouraging the development
of atherosclerotic plaques. (Reproduced with permission from [141])
antagonistic ligand to Olfr78, which may trigger SCFAs and result in hypertension
[143, 144]. There’s no better place to start than in the gut when it comes to cancer.
SCFA stimulates GPR43 to offset the effects of Olfr78-mediated renin release
[145]. GPR41 and Olfr78 may have conflicting impacts on blood pressure control
following the propionate response [146, 147]. Vasoactive oxygen species generation
is decreased, monocyte chemoattractant protein levels fall, vascular leukocyte, neu-
trophil, and monocyte adhesion to the artery wall are reduced by angiotensin II
injection in aseptic mice.
Because of this, individuals with high plasma TMAO concentrations had a higher
risk of cardiovascular events throughout 3 years [148]. TMAO levels were reported
to be increased in mouse plaque and foam cells [149, 150]. TMAO improves angio-
tensin II’s pressor action on blood pressure (Ang II). As long as TMAO and Ang II
are administered simultaneously, the pressor effect may remain throughout the trial.
TMAO alone had little impact on rat blood pressure, but it increased the pressor
effect when combined with low-dose Ang II [151]. As angiopoietin-2 or angiopoi-
etin-2 analogs are processed, TMAO may enhance the pressor activity of the recep-
tor proteins [152]. It was used to explore citrus flavonoids in a variety of cell types.
Hesperetin and naringenin both slowed the growth of apoB100 in HepG2 cells for
4 hours. If given to HepG2 cells, naringenin lowers the quantity of microsomal
transfer proteins and cholesterol acyltransferase that may be employed for lipopro-
tein formation [153–155].
Role of Polyphenols in Cardiovascular Diseases 877
To test this hypothesis, lemon peel polyphenols, such as eriodictyol and hesperi-
din, were added to a high-fat diet (HFD) in C57BL/6 mice [156]. In diabetic/dia-
betic mice (db/db), orange peel extract decreased liver fat and blood plasma levels.
In Wistar rats fed a high-cholesterol diet for 90 days, naringenin (50 mg/kg) was
shown to decrease plasma lipids and liver fat, as well as MMP gene expression and
macrophage infiltration indicators. Citrus flavonoid and tocotrienol intake reduced
total plasma cholesterol by 20–30%, LDL by 19–27%, and TG by 20–30% over
4 weeks (24–34%). For a year, people with moderate hypercholesterolemia were
shown to have lower total cholesterol, LDL cholesterol, and apoB. At 500 mg/day
for 24 weeks, individuals with hypertriglyceridemia had significant reductions in
their levels of both plasma triglycerides (TG) and apoB (apolipoprotein B). A big-
ger Japanese research found a link between frequent consumption of citrus fruits
(six to seven times weekly) and a lower risk of cardiovascular disease, especially
ischemic stroke (10,623 participants: 4147 men and 6476 women) [157]. There was
a decrease in fatty streak pattern and macrophage infiltration in rabbits fed with
naringin. Hypercholesterolemic rabbits’ endothelial cell ICAM-1 expression may
be reduced by naringin [158]. In rabbits with high plasma cholesterol, the antiath-
erogenic effects of naringin and naringenin may be due to their capacity to lower
aortic VCAM1 and MCP-1 expression. Dyslipidemia has been associated to abnor-
mally high apoB-containing lipoprotein levels [159]. To test this hypothesis, scien-
tists gave wild-type mice supplemental fat and naringin. Naringin may inhibit the
manufacture of apoB100 in HepG2 cells by activating a signalling cascade there
[160]. The antiatherogenic properties of naringenin were shown in Ldlr-/- mice
given a Western diet with 3% naringenin (w/w).
Flavones, a subtype of luteolin-rich flavonoids, work by signaling and activating
the cAMP/protein kinase A cascade, activating NO synthase, increasing endothelial
NO concentration [161]. This mechanism causes vasodilation, controlled by potas-
sium and calcium channels [162].
Inhibition of the renin-angiotensin-aldosterone system, improvement of endo-
thelial dysfunction, and regulation of smooth muscle contraction in vessels [163].
These methods work by activating NO-synthase 3, increasing plasma NO levels.
Endothelial function is improved by inhibiting endothelin-1 activity on smooth
muscle cells in arteries [164].
The antihypertensive impact of quercetin and kaempferol is reliant on NO pro-
duced in the endothelium, according to in vitro studies. Endothelial denudation sig-
nificantly reduced the vasodilator action of the two flavonols [165–167]. It may
activate potassium channels triggered by calcium and voltage-dependent potassium
channels, which is similar to quercetin’s vasodilatory impact in denuded endothe-
lium. The ability of naringenin to reduce blood pressure is attributable to both mem-
brane hyperpolarization and relaxing of vascular smooth muscle [168]. This
demonstrates naringenin’s therapeutic value in hypertension [169]. The second fla-
vanone, hesperetin, causes vasodilation through its active metabolite (hesperetin-7-0-
betaglucuronide) by boosting nitric oxide synthase adhesion and decreasing nitrous
oxide levels. Thus, NO levels rise in the plasma [170]. Vasodilation occurs when
intracellular calcium ions are reduced by inhibiting voltage-gated calcium channels
878 H. Chopra et al.
[171]. Many studies have indicated an inverse connection between flavonoid intake
and the incidence of acute myocardial infarction (AMI), and even problems after
AMI may be reduced by moderate red wine consumption [172]. Polyphenols (quer-
cetin, resveratrol) or diets rich in these chemicals (red wine or natural grape juice)
have been demonstrated to minimize cell suffering induced by myocardial ischemia
and improve contractile dysfunction after a heart attack. Following infarction, flavo-
noids protect myocardium cells by reducing oxidative stress and increasing NO
levels [172]. It has also been linked to a reduced risk of myocardial infarction in
young and middle-aged women [173].
In the case of brain ischemia, significant blood flow interruption causes meta-
bolic abnormalities. Local homeostatic disruptions (particularly ionic homeostasis,
acid–base balance) will trigger excessive production of excitatory amino acids.
These conditions enhance oxidative stress, causing neuronal death and tissue dam-
age. Flavonoids may help prevent ischemic strokes by lowering blood pressure,
increasing lipid oxidation, and improving endothelial function. Moreover, resvera-
trol may minimize the amount of injured tissue after ischemia in experimental mod-
els, presumably due to reduced lipid oxidation. Grape polyphenols may lower the
number of ischemia-affected neurons [174].
5 Stilbene in CVD
resveratrol (2.5 mg/kg/day) both decreased blood pressure more than either medica-
tion alone did [179]. Resveratrol might be used in combination with current drugs.
It was shown that resveratrol intake of 150 mg/day reduced blood pressure in indi-
viduals. Diastolic blood pressure is unaffected by resveratrol [180]. Pterostilbene’s
effect on blood pressure has been studied in a small number of human trials. There
were no significant differences in blood pressure between the two dosages of
Pterostilbene (125 mg twice a day vs. 50 mg twice a day) [181]. Resveratrol was
shown to inhibit platelet aggregation in animal and human studies [182, 183]. This
may be related to the COX-1 inhibitory effect of resveratrol (50 g/mL) on the ara-
chidonic acid pathway [184]. MAPK inhibition [185], nitric oxide/cGMP activation
[186], and phosphoinositide signaling inhibition [185] are further ways. Vascular
function, lipid accumulation, lipogenesis, and lipolysis gene regulation are all
affected by resveratrol [187, 188]. Resveratrol also lowers the apoptosis of endothe-
lial cells produced by oxidized LDL. Endothelial cells are similarly protected
against cell death by pterostilbene [189]. Pterostilbene activated AMPK and sup-
pressed mTOR in endothelial cells, resulting in cytoprotective autophagy [190].
Pterostilbene stimulated AMPK via the activation of CAMKK. TGF, TNF, IL-1, and
IL-6 have been shown to prevent atherosclerosis in mice by reducing pterostilbene’s
ability to inhibit cell proliferation and cell cycle progression in vascular smooth
muscle cells [191, 192]. Lipopolysaccharides, LDL cholesterol, and lipid peroxida-
tion in mice were reduced by piceatannol (15–45 mg/kg) [193, 194].
Cardiovascular disease has nitric oxide as an essential reactive nitrogen species
(RNS) in its pathogenesis. Although nitric oxide is a free radical because it lacks a
paired electron, this does not make it reactive. Oxidation of one of the guanidino
nitrogen atoms of L-arginine to L-citrulline in the presence of NADPH and O2 pro-
duces NO in the body [195]. It is produced by the endothelial NOS (eNOS) and is
essential for cardiovascular homeostasis (in endothelial cells, not in all cell types).
Low NO concentrations benefit the circulatory system by increasing blood flow, but
high NO concentrations have the opposite effect due to their reactive properties.
Treatment with resveratrol boosted eNOS mRNA expression in HUVEC and
HUVEC-derived EA.hy 926 cells [196]. eNOS protein expression and eNOS-
derived NO production were increased after long-term resveratrol incubation.
Resveratrol’s cardiovascular effects may be attributed to increased eNOS expres-
sion and activity [197]. Resveratrol and quercetin decreased intracellular NO and
superoxide production in oxLDL, but not in HUVECs that had been exposed to
LDL [198].
Resveratrol is an antioxidant because of the aromatic groups in its structure.
2,2′-azinobis(3-ethylbenzthiazoline-6-sulfonic acid) (ABTS), 1,1 diphenyl-2-
picrylhydrazyl (DPPH), and the hydroxyl radical may be sequestered by resveratrol,
according to research [199]. Resveratrol has been shown to reduce oxidative-stress-
induced apoptosis in PBMCs, RPE cells, rat pheochromacytoma cells, and mouse
3T3 fibroblasts, among others [200–202], because LDL oxidation is a key contribu-
tor to atherosclerosis, its antioxidant qualities may help prevent this illness.
Oxidation of LDL is prevented in vitro by resveratrol [203]. Reduced ROS produc-
tion and lipid peroxidation in atherosclerotic lesions have been shown by resveratrol
880 H. Chopra et al.
Many foods contain polyphenols, which may protect against CVD and contribute to
a healthy diet via in vivo research. Getting a hold on the underlying processes is
difficult. This is exacerbated by the fact that many in vitro studies use low quantities
of polyphenols, which have little bioavailability and are rapidly transformed into
less harmful phenols, aldehides, and salicyates. For this reason, in vitro, antioxidant
capacity tests should not be used as a predictor of intra vivo antioxidant activity
because of their low bioavailability and rapid metabolism. Understanding the phar-
macological actions of dietary polyphenols on receptor, cell signaling, and gene
expression is becoming increasingly complex. In many cases, polyphenol metabolic
products may be just as powerful as their parent compounds, and the levels neces-
sary to activate these pathways are often orders of magnitude smaller than those
required for direct antioxidant activity.
When it comes to polyphenols, pharmacology has to return to its origins.
Polyphenols’ stability, absorption, distribution, and metabolism have been over-
looked in the design of in vitro tests to determine their mechanism of action.
Polyphenols and their metabolic products in cell culture need to be tested in animals
before being used in cell culture. As a result, persistent exposure to low concentra-
tions in live organisms cannot be modeled using a single big dosage. Each approach
exposes a diverse set of mechanisms, even if the end outcome (antioxidant protec-
tion, for instance) is the same.
It is important to understand the pharmacological mechanism by which polyphe-
nols have cardiovascular effects, not only for dietary advice but also for the develop-
ment of medications and nutraceuticals that target the same pathways as polyphenols.
An example of a semisynthetic drug, Diosmin, contains hesperidin as its active
component, widely used in Europe and the United States, particularly in the treat-
ment of vein ischemia [208, 209]. Rather than an antioxidant effect, diosmin has a
venous constriction way of action. Although polyphenols are capable of a wide
range of beneficial effects in humans, the first step in properly understanding their
pharmacological potential is to move away from the direct antioxidant notion.
References
1. Pandey KB, Rizvi SI (2009) Plant polyphenols as dietary antioxidants in human health and
disease. Oxidative Med Cell Longev 2:270–278
2. Ganesan K, Xu B (2017) A critical review on polyphenols and health benefits of black soy-
beans. Nutrients 9:455
Role of Polyphenols in Cardiovascular Diseases 881
3. Cory H, Passarelli S, Szeto J, Tamez M, Mattei J (2018) The role of polyphenols in human
health and food systems: a mini-review. Front Nutr 5:87
4. Edmands WMB, Ferrari P, Rothwell JA, Rinaldi S, Slimani N, Barupal DK, Biessy C, Jenab
M, Clavel-Chapelon F, Fagherazzi G et al (2015) Polyphenol metabolome in human urine
and its association with intake of polyphenol-rich foods across European countries. Am J
Clin Nutr 102. https://doi.org/10.3945/ajcn.114.101881
5. Gardener H, Caunca MR (2018) Mediterranean diet in preventing neurodegenerative dis-
eases. Curr Nutr Rep 7:10–20
6. ICW A, PCH H (2005) Polyphenols and disease risk in epidemiologic studies… Proceedings
of the 1st international conference on polyphenols and health held in Vichy, France, November
18–21, 2004. Am J Clin Nutr 81:317S–325S
7. Tresserra-Rimbau A, Rimm EB, Medina-Remón A, Martínez-González MA, de la Torre R,
Corella D, Salas-Salvadó J, Gómez-Gracia E, Lapetra J, Arós F et al (2014) Inverse association
between habitual polyphenol intake and incidence of cardiovascular events in the PREDIMED
study. Nutr Metab Cardiovasc Dis 24. https://doi.org/10.1016/j.numecd.2013.12.014
8. Vita JA (2005) Polyphenols and cardiovascular disease: effects on endothelial and platelet
function. Am J Clin Nutr 81:292S–297S
9. Yamagata K (2019) Polyphenols regulate endothelial functions and reduce the risk of cardio-
vascular disease. Curr Pharm Des 25. https://doi.org/10.2174/1381612825666190722100504
10. Arts ICW, Hollman PCH (2005) Polyphenols and disease risk in epidemiologic studies. Am
J Clin Nutr 81:317S–325S
11. Kocic B, Kitic D, Brankovic S (2013) Dietary flavonoid intake and colorectal cancer risk:
evidence from human population studies. J BUON 18:34
12. Hollman PCH, Katan MB (1997) Absorption, metabolism and health effects of dietary fla-
vonoids in man. Biomed Pharmacother 51. https://doi.org/10.1016/S0753-3322(97)88045-6
13. Khan J, Deb PK, Priya S, Medina KD, Devi R, Walode SG, Rudrapal M (2021) Dietary fla-
vonoids: cardioprotective potential with antioxidant effects and their pharmacokinetic, toxi-
cological and therapeutic concerns. Molecules 26:4021
14. Graf BA, Milbury PE, Blumberg JB (2005) Flavonols, flavones, flavanones, and human
health: epidemiological evidence. J Med Food 8:281–290
15. Beckman CH (2000) Phenolic-storing cells: keys to programmed cell death and periderm
formation in wilt disease resistance and in general defence responses in plants? Physiol Mol
Plant Pathol 57. https://doi.org/10.1006/pmpp.2000.0287
16. Spencer JPE, Abd El Mohsen MM, Minihane AM, Mathers JC (2008) Biomarkers of the
intake of dietary polyphenols: strengths, limitations and application in nutrition research. Br
J Nutr 99:12–22
17. Rodríguez-García C, Sánchez-Quesada C, Toledo E, Delgado-Rodríguez M, Gaforio JJ
(2019) Naturally lignan-rich foods: a dietary tool for health promotion? Molecules 24:917
18. Mutha RE, Tatiya AU, Surana SJ (2021) Flavonoids as natural phenolic compounds and
their role in therapeutics: an overview. Future J Pharm Sci 7. https://doi.org/10.1186/
s43094-020-00161-8
19. Tungmunnithum D, Thongboonyou A, Pholboon A, Yangsabai A (2018) Flavonoids and
other phenolic compounds from medicinal plants for pharmaceutical and medical aspects: an
overview. Medicines 5. https://doi.org/10.3390/medicines5030093
20. Zhao J, Yang J, Xie Y (2019) Improvement strategies for the oral bioavailability of poorly
water-soluble flavonoids: an overview. Int J Pharm 570:118642
21. Kumar S, Pandey AK (2013) Chemistry and biological activities of flavonoids: an overview.
Sci World J 2013:162750
22. Panche AN, Diwan AD, Chandra SR (2016) Flavonoids: an overview. J Nutr Sci 5:e47
23. Amarowicz R, Pegg RB (2017) The potential protective effects of phenolic compounds
against low-density lipoprotein oxidation. Curr Pharm Des 23. https://doi.org/10.217
4/1381612823666170329142936
882 H. Chopra et al.
24. Bouarab Chibane L, Degraeve P, Ferhout H, Bouajila J, Oulahal N (2019) Plant antimicrobial
polyphenols as potential natural food preservatives. J Sci Food Agric 99:1457–1474
25. Leyva-Jimenez FJ, Lozano-Sanchez J, Borras-Linares I, de la Cadiz-Gurrea ML, Mahmoodi-
Khaledi E (2019) Potential antimicrobial activity of honey phenolic compounds against gram
positive and gram negative bacteria. LWT 101. https://doi.org/10.1016/j.lwt.2018.11.015
26. Aguirre-Becerra H, Pineda-Nieto SA, García-Trejo JF, Guevara-González RG, Feregrino-
Pérez AA, Álvarez-Mayorga BL, Rivera Pastrana DM (2020) Jacaranda flower (Jacaranda
mimosifolia) as an alternative for antioxidant and antimicrobial use. Heliyon 6. https://doi.
org/10.1016/j.heliyon.2020.e05802
27. Takó M, Kerekes EB, Zambrano C, Kotogán A, Papp T, Krisch J, Vágvölgyi C (2020) Plant
phenolics and phenolic-enriched extracts as antimicrobial agents against food-contaminating
microorganisms. Antioxidants 9:165
28. Mammadov R, Kaska A, Ozay C (2017) Phenolic composition, antioxidant and cyto-
toxic activities of Prospero autumnale. Indian J Pharm Sci 79. https://doi.org/10.4172/
pharmaceutical-sciences.1000266
29. Yang CS, Landau JM, Huang MT, Newmark HL (2001) Inhibition of carcinogenesis by
dietary polyphenolic compounds. Annu Rev Nutr 21:386–406
30. Friedman MC (1997) Biochemistry, and dietary role of potato polyphenols. A review. J Agric
Food Chem 45:1523–1540
31. Olivas-Quintero S, López-Angulo G, Montes-Avila J, Díaz-Camacho SP, Vega-Aviña R,
López-Valenzuela JÁ, Salazar-Salas NY, Delgado-Vargas F (2017) Chemical composition
and biological activities of Helicteres vegae and Heliopsis sinaloensis. Pharm Biol 55. https://
doi.org/10.1080/13880209.2017.1306712
32. Chariyakornkul A, Punvittayagul C, Taya S, Wongpoomchai R (2019) Inhibitory effect of
purple rice husk extract on AFB1-induced micronucleus formation in rat liver through modu-
lation of xenobiotic metabolizing enzymes. BMC Complement Altern Med 19. https://doi.
org/10.1186/s12906-019-2647-9
33. Mushtaq M, Sultana B, Anwar F, Batool S (2015) Antimutagenic and antioxidant potential of
aqueous and acidified methanol extracts from citrus limonum fruit residues. J Chil Chem Soc
60. https://doi.org/10.4067/S0717-97072015000200025
34. Beaver C, Collins TS, Harbertson J (2020) Red wine phenolic compounds using ultraviolet –
visible spectra. Molecules 25:1–8
35. Fourie E, Aleixandre-Tudo JL, Mihnea M, du Toit W (2020) Partial least squares calibra-
tions and batch statistical process control to monitor phenolic extraction in red wine fermen-
tations under different maceration conditions. Food Control 115. https://doi.org/10.1016/j.
foodcont.2020.107303
36. Fang X, Azain M, Crowe-White K, Mumaw J, Grimes JA, Schmiedt C, Barletta M, Rayalam
S, Park HJ (2019) Effect of acute ingestion of green tea extract and lemon juice on oxidative
stress and lipid profile in pigs fed a high-fat diet. Antioxidants 8. https://doi.org/10.3390/
antiox8060195
37. Ahn-Jarvis JH, Parihar A, Doseff AI (2019) Dietary flavonoids for immunoregulation and
cancer: food design for targeting disease. Antioxidants 8:202
38. Buttar HS, Li T, Ravi N (2005) Prevention of cardiovascular diseases: role of exercise, dietary
interventions, obesity and smoking cessation. Exp Clin Cardiol 10:229–249
39. Katz DL, Doughty K, Ali A (2011) Cocoa and chocolate in human health and disease.
Antioxid Redox Signal 15:2779–2811
40. Huxley RR, Neil HAW (2003) The relation between dietary flavonol intake and coronary heart
disease mortality: a meta-analysis of prospective cohort studies. Eur J Clin Nutr 57:904–908
41. Lagiou P, Samoli E, Lagiou A, Tzonou A, Kalandidi A, Peterson J, Dwyer J, Trichpoulos D
(2004) Intake of specific flavonoid classes and coronary heart disease – a case-control study
in Greece. Eur J Clin Nutr 58. https://doi.org/10.1038/sj.ejcn.1602022
Role of Polyphenols in Cardiovascular Diseases 883
42. Rimm EB, Katan MB, Ascherio A, Stampfer MJ, Willett WC (1996) Relation between intake
of flavonoids and risk for coronary heart disease in male health professionals. Ann Intern
Med 125. https://doi.org/10.7326/0003-4819-125-5-199609010-00005
43. Hirvonen T, Pietinen P, Virtanen M, Ovaskainen ML, Häkkinen S, Albanes D, Virtamo J
(2001) Intake of flavonols and flavones and risk of coronary heart disease in male smokers.
Epidemiology 12. https://doi.org/10.1097/00001648-200101000-00011
44. Bondonno NP, Dalgaard F, Kyrø C, Murray K, Bondonno CP, Lewis JR, Croft KD, Gislason
G, Scalbert A, Cassidy A et al (2019) Flavonoid intake is associated with lower mortal-
ity in the Danish diet cancer and health cohort. Nat Commun 10. https://doi.org/10.1038/
s41467-019-11622-x
45. Peterson JJ, Dwyer JT, Jacques PF, McCullough ML (2012) Do flavonoids reduce cardiovas-
cular disease incidence or mortality in US and European populations? Nutr Rev 70:491–508
46. Kruger MJ, Davies N, Myburgh KH, Lecour S (2014) Proanthocyanidins, anthocyanins and
cardiovascular diseases. Food Res Int 59:41–52
47. Wallace TC (2011) Anthocyanins in cardiovascular disease. Adv Nutr 2:1–7
48. Gardner EJ, Ruxton CHS, Leeds AR (2007) Black tea – helpful or harmful? A review of the
evidence. Eur J Clin Nutr 61:3–18. https://doi.org/10.1038/SJ.EJCN.1602489
49. Li D, Wang R, Huang J, Cai Q, Yang CS, Wan X, Xie Z (2019) Effects and mechanisms of tea
regulating blood pressure: evidences and promises. Nutrients 11:E1115
50. Igho-Osagie E, Cara K, Wang D, Yao Q, Penkert LP, Cassidy A, Ferruzzi M, Jacques PF,
Johnson EJ, Chung M et al (2020) Short-term tea consumption is not associated with a reduc-
tion in blood lipids or pressure: a systematic review and meta-analysis of randomized con-
trolled trials. J Nutr 150. https://doi.org/10.1093/jn/nxaa295
51. Tuteja N, Chandra M, Tuteja R, Misra MK (2004) Nitric oxide as a unique bioactive signaling
messenger in physiology and pathophysiology. J Biomed Biotechnol 2004:227–237
52. Förstermann U, Sessa WC (2012) Nitric oxide synthases: regulation and function. Eur Heart
J 33:829–837
53. Blumberg JB, Vita JA, Oliver Chen CY (2015) Concord grape juice polyphenols and car-
diovascular risk factors: dose-response relationships. Nutrients 7. https://doi.org/10.3390/
nu7125519
54. Duffy SJ, Vita JA, Holbrook M, Swerdloff PL, Keaney JF (2001) Effect of acute and chronic
tea consumption on platelet aggregation in patients with coronary artery disease. Arterioscler
Thromb Vasc Biol 21. https://doi.org/10.1161/01.ATV.21.6.1084
55. McEwen BJ (2014) The influence of diet and nutrients on platelet function. Semin Thromb
Hemost 40. https://doi.org/10.1055/s-0034-1365839
56. Rees A, Dodd GF, Spencer JPE (2018) The effects of flavonoids on cardiovascular health:
a review of human intervention trials and implications for cerebrovascular function.
Nutrients 10:1852
57. Peters U, Poole C, Arab L (2001) Does tea affect cardiovascular disease? A meta-analysis.
Am J Epidemiol 154. https://doi.org/10.1093/aje/154.6.495
58. Tresserra-Rimbau A, Rimm EB, Medina-Remón A, Martínez-González MA, López-
Sabater MC, Covas MI, Corella D, Salas-Salvadó J, Gómez-Gracia E, Lapetra J et al (2014)
Polyphenol intake and mortality risk: a re-analysis of the PREDIMED trial. BMC Med 12.
https://doi.org/10.1186/1741-7015-12-77
59. Tresserra-Rimbau A, Medina-Remón A, Pérez-Jiménez J, Martínez-González MA, Covas
MI, Corella D, Salas-Salvadó J, Gómez-Gracia E, Lapetra J, Arós F et al (2013) Dietary
intake and major food sources of polyphenols in a Spanish population at high cardiovascu-
lar risk: the PREDIMED study. Nutr Metab Cardiovasc Dis 23. https://doi.org/10.1016/j.
numecd.2012.10.008
60. Sarwar N, Gao P, Kondapally Seshasai SR, Gobin R, Kaptoge S, Di Angelantonio E,
Ingelsson E, Lawlor DA, Selvin E, Stampfer M et al (2010) Diabetes mellitus, fasting blood
glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102
884 H. Chopra et al.
77. Warner TD, Nylander S, Whatling C (2011) Anti-platelet therapy: cyclo-oxygenase inhi-
bition and the use of aspirin with particular regard to dual anti-platelet therapy. Br J Clin
Pharmacol 72:619–633
78. Mattiello T, Trifirò E, Jotti GS, Pulcinelli FM (2009) Effects of pomegranate juice and extract
polyphenols on platelet function. J Med Food 12. https://doi.org/10.1089/jmf.2007.0640
79. Karlíčková J, Říha M, Filipský T, Macáková K, Hrdina R, Mladěnka P (2015) Antiplatelet
effects of flavonoids mediated by inhibition of arachidonic acid based pathway. Planta Med
82. https://doi.org/10.1055/s-0035-1557902
80. Wu CM, Wu SC, Chung WJ, Lin HC, Chen KT, Chen YC, Hsu MF, Yang JM, Wang JP, Lin
CN (2007) Antiplatelet effect and selective binding to cyclooxygenase (COX) by molecular
docking analysis of flavonoids and lignans. Int J Mol Sci 8. https://doi.org/10.3390/i8080830
81. Calixto NO, E Silva MCDC, Gayer CRM, Coelho MGP, Paes MC, Todeschini AR (2007)
Antiplatelet activity of geranylgeraniol isolated from Pterodon pubescens fruit oil is mediated
by inhibition of cyclooxygenase-1. Planta Med 73. https://doi.org/10.1055/s-2007-967177
82. Nurtjahja-Tjendraputra E, Ammit AJ, Roufogalis BD, Tran VH, Duke CC (2003) Effective
anti-platelet and COX-1 enzyme inhibitors from pungent constituents of ginger. Thromb Res
111. https://doi.org/10.1016/j.thromres.2003.09.009
83. Bijak M, Saluk-Bijak J (2017) Flavonolignans inhibit the arachidonic acid pathway in blood
platelets. BMC Complement Altern Med 17. https://doi.org/10.1186/s12906-017-1897-7
84. Chang MC, Chang HH, Chan CP, Chou HY, Chang BE, Yeung SY, Wang TM, Jeng JH (2012)
Antiplatelet effect of phloroglucinol is related to inhibition of cyclooxygenase, reactive oxy-
gen species, ERK/p38 signaling and thromboxane A2 production. Toxicol Appl Pharmacol
263:287–295. https://doi.org/10.1016/J.TAAP.2012.06.021
85. Applová L, Karlíčková J, Říha M, Filipský T, Macáková K, Spilková J, Mladěnka P (2017)
The isoflavonoid tectorigenin has better antiplatelet potential than acetylsalicylic acid.
Phytomedicine 35. https://doi.org/10.1016/j.phymed.2017.08.023
86. Guerrero JA, Lozano ML, Castillo J, Benavente-García O, Vicente V, Rivera J (2005)
Flavonoids inhibit platelet function through binding to the thromboxane A2 receptor. J
Thromb Haemost 3. https://doi.org/10.1111/j.1538-7836.2004.01099.x
87. Lee JJ, Jin YR, Lim Y, Hong JT, Kim TJ, Chung JH, Yun YP (2006) Antiplatelet activity of
carnosol is mediated by the inhibition of TXA2 receptor and cytosolic calcium mobilization.
Vasc Pharmacol 45. https://doi.org/10.1016/j.vph.2006.04.003
88. Srivastava KC, Tyagi OD (1993) Effects of a garlic-derived principle (ajoene) on aggregation
and arachidonic acid metabolism in human blood platelets. Prostaglandins Leukot Essent Fat
Acids 49. https://doi.org/10.1016/0952-3278(93)90165-S
89. Ju HK, Lee JG, Park MK, Park SJ, Lee CH, Park JH, Kwon SW (2012) Metabolomic
investigation of the anti-platelet aggregation activity of ginsenoside Rk1 reveals attenuated
12-HETE production. J Proteome Res 11. https://doi.org/10.1021/pr300454f
90. Hanssen NMJ, Wouters K, Huijberts MS, Gijbels MJ, Sluimer JC, Scheijen JLJM, Heeneman
S, Biessen EAL, Daemen MJAP, Brownlee M et al (2014) Higher levels of advanced gly-
cation endproducts in human carotid atherosclerotic plaques are associated with a rupture-
prone phenotype. Eur Heart J 35. https://doi.org/10.1093/eurheartj/eht402
91. Rabbani N, Thornalley PJ (2018) Advanced glycation end products in the pathogenesis of
chronic kidney disease. Kidney Int 93:803–813
92. Lo CY, Hsiao WT, Chen XY (2011) Efficiency of trapping methylglyoxal by phenols and
phenolic acids. J Food Sci 76. https://doi.org/10.1111/j.1750-3841.2011.02067.x
93. Totlani VM, Peterson DG (2006) Epicatechin carbonyl-trapping reactions in aqueous
Maillard systems: identification and structural elucidation. J Agric Food Chem 54. https://
doi.org/10.1021/jf061244r
94. Vlassara H, Cai W, Tripp E, Pyzik R, Yee K, Goldberg L, Tansman L, Chen X, Mani V,
Fayad ZA et al (2016) Oral AGE restriction ameliorates insulin resistance in obese individu-
als with the metabolic syndrome: a randomised controlled trial. Diabetologia 59. https://doi.
org/10.1007/s00125-016-4053-x
886 H. Chopra et al.
95. Baye E, Kiriakova V, Uribarri J, Moran LJ, De Courten B (2017) Consumption of diets with
low advanced glycation end products improves cardiometabolic parameters: meta-analysis of
randomised controlled trials. Sci Rep 7. https://doi.org/10.1038/s41598-017-02268-0
96. Rodríguez JM, Leiva Balich L, Concha MJ, Mizón C, Bunout Barnett D, Barrera Acevedo
G, Hirsch Birn S, Jiménez Jaime T, Henríquez S, Uribarri J et al (2015) Reduction of serum
advanced glycation end-products with a low calorie Mediterranean diet. Nutr Hosp 31.
https://doi.org/10.3305/nh.2015.31.6.8936
97. Lopez-Moreno J, Quintana-Navarro GM, Delgado-Lista J, Garcia-Rios A, Alcala-Diaz JF,
Gomez-Delgado F, Camargo A, Perez-Martinez P, Tinahones FJ, Striker GE et al (2018)
Mediterranean diet supplemented with coenzyme Q 10 modulates the postprandial metabo-
lism of advanced glycation end products in elderly men and women. J Gerontol A Biol Sci
Med Sci 73. https://doi.org/10.1093/gerona/glw214
98. Urquiaga I, Ávila F, Echeverria G, Perez D, Trejo S, Leighton F (2017) A Chilean berry concen-
trate protects against postprandial oxidative stress and increases plasma antioxidant activity
in healthy humans. Oxidative Med Cell Longev 2017. https://doi.org/10.1155/2017/8361493
99. Shao B, Pennathur S, Pagani I, Oda MN, Witztum JL, Oram JF, Heinecke JW (2010)
Modifying apolipoprotein A-I by malondialdehyde, but not by an array of other reactive
carbonyls, blocks cholesterol efflux by the ABCA1 pathway. J Biol Chem 285. https://doi.
org/10.1074/jbc.M110.118182
100. Pamplona R, Portero-Otín M, Riba D, Requena JR, Thorpe SR, López-Torres M, Barja G
(2000) Low fatty acid unsaturation: a mechanism for lowered lipoperoxidative modification
of tissue proteins in mammalian species with long life spans. J Gerontol A Biol Sci Med Sci
55. https://doi.org/10.1093/gerona/55.6.B286
101. Ruiz MC, Ayala V, Portero-Otín M, Requena JR, Barja G, Pamplona R (2005) Protein methi-
onine content and MDA-lysine adducts are inversely related to maximum life span in the
heart of mammals. Mech Ageing Dev 126. https://doi.org/10.1016/j.mad.2005.04.005
102. Ramkissoon JS, Mahomoodally MF, Ahmed N, Subratty AH (2013) Antioxidant and anti-
glycation activities correlates with phenolic composition of tropical medicinal herbs. Asian
Pac J Trop Med 6. https://doi.org/10.1016/S1995-7645(13)60097-8
103. Harris CS, Cuerrier A, Lamont E, Haddad PS, Arnason JT, Bennett SAL, Johns T (2014)
Investigating wild berries as a dietary approach to reducing the formation of advanced glyca-
tion endproducts: chemical correlates of in vitro antiglycation activity. Plant Foods Hum Nutr
69. https://doi.org/10.1007/s11130-014-0403-3
104. Kokkinidou S, Peterson DG (2013) Response surface methodology as optimization strategy
for reduction of reactive carbonyl species in foods by means of phenolic chemistry. Food
Funct 4. https://doi.org/10.1039/c3fo60032g
105. Li X, Zheng T, Sang S, Lv L (2014) Quercetin inhibits advanced glycation end product forma-
tion by trapping methylglyoxal and glyoxal. J Agric Food Chem 62. https://doi.org/10.1021/
jf504132x
106. Lv L, Shao XI, Wang L, Huang D, Ho CT, Sang S (2010) Stilbene glucoside from polygonum
multiflorum thunb.: a novel natural inhibitor of advanced glycation end product formation by
trapping of methylglyoxal. J Agric Food Chem 58. https://doi.org/10.1021/jf904122q
107. Shen Y, Xu Z, Sheng Z (2017) Ability of resveratrol to inhibit advanced glycation end product
formation and carbohydrate-hydrolyzing enzyme activity, and to conjugate methylglyoxal.
Food Chem 216. https://doi.org/10.1016/j.foodchem.2016.08.034
108. Van Den Eynde MDG, Geleijnse JM, Scheijen JLJM, Hanssen NMJ, Dower JI, Afman LA,
Stehouwer CDA, Hollman PCH, Schalkwijk CG (2018) Quercetin, but not epicatechin,
decreases plasma concentrations of methylglyoxal in adults in a randomized, double-blind,
placebo-controlled, crossover trial with pure flavonoids. J Nutr 148. https://doi.org/10.1093/
jn/nxy236
109. Dower JI, Geleijnse JM, Gijsbers L, Zock PL, Kromhout D, Hollman PCH (2015) Effects
of the pure flavonoids epicatechin and quercetin on vascular function and cardiometabolic
health: a randomized, double-blind, placebo-controlled, crossover trial. Am J Clin Nutr 101.
https://doi.org/10.3945/ajcn.114.098590
Role of Polyphenols in Cardiovascular Diseases 887
110. Zordoky BNM, Robertson IM, Dyck JRB (2014) Preclinical and clinical evidence for the role
of resveratrol in the treatment of cardiovascular diseases. Biochim Biophys Acta Mol basis
Dis 1852:1155–1177
111. Zhang N, Wei WY, Li LL, Hu C, Tang QZ (2018) Therapeutic potential of polyphenols in
cardiac fibrosis. Front Pharmacol 9:122
112. Han X, Gao S, Cheng Y, Sun Y, Liu W, Tang L, Ren D (2012) Protective effect of naringenin-7-
O-glucoside against oxidative stress induced by doxorubicin in H9c2 cardiomyocytes. Biosci
Trends 6. https://doi.org/10.5582/bst.2012.v6.1.19
113. Repo-Carrasco-Valencia R, Hellström JK, Pihlava JM, Mattila PH (2010) Flavonoids and
other phenolic compounds in Andean indigenous grains: Quinoa (Chenopodium quinoa),
kañiwa (Chenopodium pallidicaule) and kiwicha (Amaranthus caudatus). Food Chem 120.
https://doi.org/10.1016/j.foodchem.2009.09.087
114. Razavi-Azarkhiavi K, Iranshahy M, Sahebkar A, Shirani K, Karimi G (2016) The protective
role of phenolic compounds against doxorubicin-induced cardiotoxicity: a comprehensive
review. Nutr Cancer 68:892–917
115. Sun J, Sun G, Meng X, Wang H, Luo Y, Qin M, Ma B, Wang M, Cai D, Guo P et al (2013)
Isorhamnetin protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS
One 8. https://doi.org/10.1371/journal.pone.0064526
116. Korga A, Józefczyk A, Zgórka G, Homa M, Ostrowska M, Burdan F, Dudka J (2017)
Evaluation of the phytochemical composition and protective activities of methanolic extracts
of Centaurea borysthenica and Centaurea daghestanica (Lipsky) Wagenitz on cardiomyocytes
treated with doxorubicin. Food Nutr Res 61. https://doi.org/10.1080/16546628.2017.134407
7
117. Alhaider IA, Mohamed ME, Ahmed KKM, Kumar AHS (2017) Date palm (Phoenix dac-
tylifera) fruits as a potential cardioprotective agent: the role of circulating progenitor cells.
Front Pharmacol 8:592. https://doi.org/10.3389/FPHAR.2017.00592/BIBTEX
118. Syama HP, Arya AD, Dhanya R, Nisha P, Sundaresan A, Jacob E, Jayamurthy P (2017)
Quantification of phenolics in Syzygium cumini seed and their modulatory role on tertiary
butyl-hydrogen peroxide-induced oxidative stress in H9c2 cell lines and key enzymes in car-
dioprotection. J Food Sci Technol 54. https://doi.org/10.1007/s13197-017-2651-3
119. Garjani A, Tila D, Hamedeyazdan S, Vaez H, Rameshrad M, Pashaii M, Fathiazad F (2017)
An investigation on cardioprotective potential of Marrubium vulgare aqueous fraction against
ischaemia-reperfusion injury in isolated rat heart. Folia Morphol (Warsz) 76. https://doi.
org/10.5603/FM.a2017.0011
120. Dludla PV, Joubert E, Muller CJF, Louw J, Johnson R (2017) Hyperglycemia-induced oxida-
tive stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic
acid-2-O-β-D-glucoside. Nutr Metab 14:45
121. Tian J, Wu X, Zhang M, Zhou Z, Liu Y (2018) Comparative study on the effects of apple peel
polyphenols and apple flesh polyphenols on cardiovascular risk factors in mice. Clin Exp
Hypertens 40. https://doi.org/10.1080/10641963.2017.1313851
122. Piano MR (2017) Alcohol’s effects on the cardiovascular system. Alcohol Res 38:219–241
123. Wilsnack RW, Wilsnack SC, Kristjanson AF, Vogeltanz-Holm ND, Gmel G (2009) Gender
and alcohol consumption: patterns from the multinational GENACIS project. Addiction 104.
https://doi.org/10.1111/j.1360-0443.2009.02696.x
124. Rimm EB, Williams P, Fosher K, Criqui M, Stampfer MJ (1999) Moderate alcohol intake
and lower risk of coronary heart disease: meta-analysis of effects on lipids and haemostatic
factors. Br Med J 319. https://doi.org/10.1136/bmj.319.7224.1523
125. Thadhani R, Camargo CA, Stampfer MJ, Curhan GC, Willett WC, Rimm EB (2002)
Prospective study of moderate alcohol consumption and risk of hypertension in young
women. Arch Intern Med 162. https://doi.org/10.1001/archinte.162.5.569
126. Leger ASS, Cochrane AL, Moore F (1979) Factors associated with cardiac mortality in devel-
oped countries with particular reference to the consumption of wine. Lancet 313. https://doi.
org/10.1016/S0140-6736(79)92765-X
888 H. Chopra et al.
127. Renaud S, de Lorgeril M (1992) Wine, alcohol, platelets, and the French paradox for coro-
nary heart disease. Lancet 339. https://doi.org/10.1016/0140-6736(92)91277-F
128. Lippi G, Franchini M, Favaloro EJ, Targher G (2010) Moderate red wine consumption and
cardiovascular disease risk: beyond the French paradox. Semin Thromb Hemost 36:59–70
129. Haseeb S, Alexander B, Baranchuk A, Electrophysiology C (2017) Wine and cardiovascular
health a comprehensive review in depth. Circulation 136:1434–1448
130. Ditano-Vázquez P, Torres-Peña JD, Galeano-Valle F, Pérez-Caballero AI, Demelo-Rodríguez
P, Lopez-Miranda J, Katsiki N, Delgado-Lista J, Alvarez-Sala-Walther LA (2019) The fluid
aspect of the mediterranean diet in the prevention and management of cardiovascular disease
and diabetes: the role of polyphenol content in moderate consumption of wine and olive oil.
Nutrients 11:2833
131. Castaldo L, Narváez A, Izzo L, Graziani G, Gaspari A, Minno G, Di; Ritieni, A. (2019) Red
wine consumption and cardiovascular health. Molecules 24:3626
132. Hansen AS, Marckmann P, Dragsted LO, Finné Nielsen IL, Nielsen SE, Grønbæk M (2005)
Effect of red wine and red grape extract on blood lipids, haemostatic factors, and other risk
factors for cardiovascular disease. Eur J Clin Nutr 59. https://doi.org/10.1038/sj.ejcn.1602107
133. Truelsen T, Grønbæk M, Schnohr P, Boysen G (1998) Intake of beer, wine, and spirits and
risk of stroke: the Copenhagen City heart study. Stroke 29. https://doi.org/10.1161/01.
STR.29.12.2467
134. Gronbaek M, Becker U, Johansen D, Gottschau A, Schnohr P, Hein HO, Jensen G, Sorensen
TIA (2000) Type of alcohol consumed and mortality from all causes, coronary heart disease,
and cancer. Ann Intern Med 133. https://doi.org/10.7326/0003-4819-133-6-200009190-00008
135. Klatsky AL (1999) Moderate drinking and reduced risk of heart disease. Alcohol Res Health
23:15–23
136. Mostofsky E, Mukamal KJ, Giovannucci EL, Stampfer MJ, Rimm EB (2016) Key findings
on alcohol consumption and a variety of health outcomes from the nurses’ health study. Am
J Public Health 106:1586–1591
137. Pingitore A, Chambers ES, Hill T, Maldonado IR, Liu B, Bewick G, Morrison DJ, Preston T,
Wallis GA, Tedford C et al (2017) The diet-derived short chain fatty acid propionate improves
beta-cell function in humans and stimulates insulin secretion from human islets in vitro.
Diabetes Obes Metab 19. https://doi.org/10.1111/dom.12811
138. Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, Bultman SJ (2011)
The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian
colon. Cell Metab 13. https://doi.org/10.1016/j.cmet.2011.02.018
139. Romano KA, Vivas EI, Amador-Noguez D, Rey FE (2015) Intestinal microbiota composition
modulates choline bioavailability from diet and accumulation of the proatherogenic metabo-
lite trimethylamine-N-oxide. MBio 6. https://doi.org/10.1128/mBio.02481-14
140. Roberts AB, Gu X, Buffa JA, Hurd AG, Wang Z, Zhu W, Gupta N, Skye SM, Cody DB,
Levison BS et al (2018) Development of a gut microbe–targeted nonlethal therapeutic to
inhibit thrombosis potential. Nat Med 24. https://doi.org/10.1038/s41591-018-0128-1
141. Li Q, Gao B, Siqin B, He Q, Zhang R, Meng X, Zhang N, Zhang N, Li M (2021) Gut micro-
biota: a novel regulator of cardiovascular disease and key factor in the therapeutic effects of
flavonoids. Front Pharmacol 12. https://doi.org/10.3389/fphar.2021.651926
142. Wilson Tang WH, Hazen SL (2017) The gut microbiome and its role in cardiovascular dis-
eases. Circulation 135. https://doi.org/10.1161/CIRCULATIONAHA.116.024251
143. Pluznick JL (2017) Microbial short-chain fatty acids and blood pressure regulation. Curr
Hypertens Rep 19:25
144. Natarajan N, Hori D, Flavahan S, Steppan J, Flavahan NA, Berkowitz DE, Pluznick
JL (2016) Microbial short chain fatty acid metabolites lower blood pressure via endo-
thelial G protein-coupled receptor 41. Physiol Genomics 48. https://doi.org/10.1152/
physiolgenomics.00089.2016
145. Chiou VL, Burotto M (2015) Pseudoprogression and immune-related response in solid
tumors. J Clin Oncol 33:3541–3543
Role of Polyphenols in Cardiovascular Diseases 889
146. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, Rey F,
Wang T et al (2013) Olfactory receptor responding to gut microbiotaderived signals plays a
role in renin secretion and blood pressure regulation. Proc Natl Acad Sci U S A 110. https://
doi.org/10.1073/pnas.1215927110
147. Jia Q, Xie Y, Lu C, Zhang A, Lu Y, Lv S, Zhang J (2019) Endocrine organs of cardiovascular
diseases: gut microbiota. J Cell Mol Med 23:2314–2323
148. Tang TWH, Chen HC, Chen CY, Yen CYT, Lin CJ, Prajnamitra RP, Chen LL, Ruan
SC, Lin JH, Lin PJ et al (2019) Loss of gut microbiota alters immune system composi-
tion and cripples postinfarction cardiac repair. Circulation 139. https://doi.org/10.1161/
CIRCULATIONAHA.118.035235
149. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu
X, Chung YM et al (2011) Gut flora metabolism of phosphatidylcholine promotes cardiovas-
cular disease. Nature 472. https://doi.org/10.1038/nature09922
150. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L
et al (2013) Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes
atherosclerosis. Nat Med 19. https://doi.org/10.1038/nm.3145
151. Ufnal M, Jazwiec R, Dadlez M, Drapala A, Sikora M, Skrzypecki J (2014) Trimethylamine-
N-oxide: a carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin
II in rats. Can J Cardiol 30. https://doi.org/10.1016/j.cjca.2014.09.010
152. Wu H, Kim M, Han J (2016) Icariin metabolism by human intestinal microflora. Molecules
21. https://doi.org/10.3390/molecules21091158
153. Borradaile NM, De Dreu LE, Huff MW (2003) Inhibition of net HepG2 cell apolipoprotein
B secretion by the citrus flavonoid naringenin involves activation of phosphatidylinositol
3-kinase, independent of insulin receptor substrate-1 phosphorylation. Diabetes 52. https://
doi.org/10.2337/diabetes.52.10.2554
154. Wilcox LJ, Borradaile NM, De Dreu LE, Huff MW (2001) Secretion of hepatocyte apoB is
inhibited by the flavonoids, naringenin and hesperetin, via reduced activity and expression of
ACAT2 and MTP. J Lipid Res 42. https://doi.org/10.1016/s0022-2275(20)31634-5
155. Mulvihill EE, Allister EM, Sutherland BG, Telford DE, Sawyez CG, Edwards JY, Markle
JM, Hegele RA, Huff MW (2009) Naringenin prevents dyslipidemia, apolipoprotein B over-
production, and hyperinsulinemia in LDL receptor-null mice with diet-induced insulin resis-
tance. Diabetes 58. https://doi.org/10.2337/db09-0634
156. Fukuchi Y, Hiramitsu M, Okada M, Hayashi S, Nabeno Y, Osawa T, Naito M (2008) Lemon
polyphenols suppress diet-induced obesity by up-regulation of mRNA levels of the enzymes
involved in β-oxidation in mouse white adipose tissue. J Clin Biochem Nutr 43. https://doi.
org/10.3164/jcbn.2008066
157. Yamada T, Hayasaka S, Shibata Y, Ojima T, Saegusa T, Gotoh T, Ishikawa S, Nakamura
Y, Kayaba K (2011) Frequency of citrus fruit intake is associated with the incidence of
cardiovascular disease: the Jichi Medical School cohort study. J Epidemiol 21. https://doi.
org/10.2188/jea.JE20100084
158. Choe SC, Kim HS, Jeong TS, Bok SH, Park YB (2001) Naringin has an antiatherogenic effect
with the inhibition of intercellular adhesion molecule-1 in hypercholesterolemic rabbits. J
Cardiovasc Pharmacol 38. https://doi.org/10.1097/00005344-200112000-00017
159. Vergès B (2015) Pathophysiology of diabetic dyslipidaemia: where are we? Diabetologia
58:886–899
160. Allister EM, Mulvihill EE, Barrett PHR, Edwards JY, Carter LP, Huff MW (2008) Inhibition
of apoB secretion from HepG2 cells by insulin is amplified by naringenin, independent of the
insulin receptor. J Lipid Res 49. https://doi.org/10.1194/jlr.M800297-JLR200
161. Ciumărnean L, Milaciu MV, Runcan O, Vesa SC, Răchisan AL, Negrean V, Perné MG, Donca
VI, Alexescu TG, Para I et al (2020) The effects of flavonoids in cardiovascular diseases.
Molecules 25:4320
162. Scholz EP, Zitron E, Katus HA, Karle CA (2010) Cardiovascular ion channels as a molecular
target of flavonoids. Cardiovasc Ther 28:e46–e52
890 H. Chopra et al.
181. Riche DM, Riche KD, Blackshear CT, McEwen CL, Sherman JJ, Wofford MR, Griswold
ME (2014) Pterostilbene on metabolic parameters: A randomized, double-blind,
and placebo- controlled trial. Evid Based Complement Altern Med 2014. https://doi.
org/10.1155/2014/459165
182. Olas B, Wachowicz B, Saluk-Juszczak J, Zieliński T (2002) Effect of resveratrol, a natural
polyphenolic compound, on platelet activation induced by endotoxin or thrombin. Thromb
Res 107. https://doi.org/10.1016/S0049-3848(02)00273-6
183. Stef G, Csiszar A, Lerea K, Ungvari Z, Veress G (2006) Resveratrol inhibits aggregation of
platelets from high-risk cardiac patients with aspirin resistance. J Cardiovasc Pharmacol 48.
https://doi.org/10.1097/01.fjc.0000238592.67191.ab
184. Zbikowska HM, Olas B, Wachowicz B, Krajewski T (1999) Response of blood platelets to
resveratrol. Platelets 10. https://doi.org/10.1080/09537109976103
185. Olas B, Wachowicz B, Holmsen H, Fukami MH (2005) Resveratrol inhibits polyphos-
phoinositide metabolism in activated platelets. Biochim Biophys Acta Biomembr 1714.
https://doi.org/10.1016/j.bbamem.2005.06.008
186. Shen MY, Hsiao G, Liu CL, Fong TH, Lin KH, Chou DS, Sheu JR (2007) Inhibitory mecha-
nisms of resveratrol in platelet activation: pivotal roles of p38 MAPK and NO/cyclic GMP. Br
J Haematol 139. https://doi.org/10.1111/j.1365-2141.2007.06788.x
187. Ou HC, Chou FP, Sheen HM, Lin TM, Yang CH, Huey-Herng Sheu W (2006) Resveratrol, a
polyphenolic compound in red wine, protects against oxidized LDL-induced cytotoxicity in
endothelial cells. Clin Chim Acta 364. https://doi.org/10.1016/j.cccn.2005.06.018
188. Voloshyna I, Hai O, Littlefield MJ, Carsons S, Reiss AB (2013) Resveratrol mediates anti-
atherogenic effects on cholesterol flux in human macrophages and endothelium via PPARγ
and adenosine. Eur J Pharmacol 698. https://doi.org/10.1016/j.ejphar.2012.08.024
189. Zhang L, Zhou GZ, Song W, Tan XR, Guo YQ, Zhou B, Jing H, Zhao SJ, Chen LK
(2012) Pterostilbene protects vascular endothelial cells against oxidized low-density
lipoprotein-induced apoptosis in vitro and in vivo. Apoptosis 17. https://doi.org/10.1007/
s10495-011-0653-6
190. Zhang L, Cui LQ, Zhou GZ, Jing HJ, Guo YQ, Sun WK (2013) Pterostilbene, a natural small-
molecular compound, promotes cytoprotective macroautophagy in vascular endothelial cells.
J Nutr Biochem 24. https://doi.org/10.1016/j.jnutbio.2012.06.008
191. Zhang Y, Zhang Y (2016) Pterostilbene, a novel natural plant conduct, inhibits high fat-induced
atherosclerosis inflammation via NF-κB signaling pathway in toll-like receptor 5 (TLR5)
deficient mice. Biomed Pharmacother 81. https://doi.org/10.1016/j.biopha.2016.04.031
192. Park ES, Lim Y, Hong JT, Yoo HS, Lee CK, Pyo MY, Yun YP (2010) Pterostilbene, a natural
dimethylated analog of resveratrol, inhibits rat aortic vascular smooth muscle cell prolif-
eration by blocking Akt-dependent pathway. Vasc Pharmacol 53. https://doi.org/10.1016/j.
vph.2010.04.001
193. Llarena M, Andrade F, Hasnaoui M, Portillo MP, Pérez-Matute P, Arbones-Mainar JM,
Hijona E, Villanueva-Millán MJ, Aguirre L, Carpéné C et al (2016) Potential renoprotective
effects of piceatannol in ameliorating the early-stage nephropathy associated with obesity in
obese Zucker rats. J Physiol Biochem 72. https://doi.org/10.1007/s13105-015-0457-1
194. Uchida-Maruki H, Inagaki H, Ito R, Kurita I, Sai M, Ito T (2015) Piceatannol lowers the blood
glucose level in diabetic mice. Biol Pharm Bull 38. https://doi.org/10.1248/bpb.b15-00009
195. Palmer RM (1993) The L-arginine: nitric oxide pathway. Curr Opin Nephrol Hypertens
2:122–128
196. Wallerath T, Deckert G, Ternes T, Anderson H, Li H, Witte K, Förstermann U (2002)
Resveratrol, a polyphenolic phytoalexin present in red wine, enhances expression and
activity of endothelial nitric oxide synthase. Circulation 106. https://doi.org/10.1161/01.
CIR.0000029925.18593.5C
197. Li H, Wallerath T, Förstermann U (2002) Physiological mechanisms regulating the expres-
sion of endothelial-type NO synthase. Nitric Oxide Biol Chem 7:132–147
892 H. Chopra et al.
198. Kostyuk VA, Potapovich AI, Suhan TO, De Luca C, Korkina LG (2011) Antioxidant and
signal modulation properties of plant polyphenols in controlling vascular inflammation. Eur
J Pharmacol 658. https://doi.org/10.1016/j.ejphar.2011.02.022
199. Soares DG, Andreazza AC, Salvador M (2003) Sequestering ability of butylated hydroxy-
toluene, propyl gallate, resveratrol, and vitamins C and E against ABTS, DPPH, and
hydroxyl free radicals in chemical and biological systems. J Agric Food Chem 51. https://
doi.org/10.1021/jf020864z
200. King RE, Kent KD, Bomser JA (2005) Resveratrol reduces oxidation and proliferation of
human retinal pigment epithelial cells via extracellular signal-regulated kinase inhibition.
Chem Biol Interact 151. https://doi.org/10.1016/j.cbi.2004.11.003
201. Chan CM, Huang CH, Li HJ, Hsiao CY, Su CC, Lee PL, Hung CF (2015) Protective effects
of resveratrol against UVA-induced damage in ARPE19 cells. Int J Mol Sci 16. https://doi.
org/10.3390/ijms16035789
202. Neal SE, Buehne KL, Besley NA, Yang P, Silinski P, Hong J, Ryde IT, Meyer JN, Jaffe GJ
(2020) Resveratrol protects against hydroquinone-induced oxidative threat in retinal pigment
epithelial cells. Investig Ophthalmol Vis Sci 61. https://doi.org/10.1167/iovs.61.4.32
203. Losa GA (2003) Resveratrol modulates apoptosis and oxidation in human blood mononu-
clear cells. Eur J Clin Investig 33. https://doi.org/10.1046/j.1365-2362.2003.01219.x
204. Guo R, Su Y, Liu B, Li S, Zhou S, Xu Y (2014) Resveratrol suppresses oxidised low-density
lipoprotein-induced macrophage apoptosis through inhibition of intracellular reactive oxygen
species generation, lox-1, and the p38 MAPK pathway. Cell Physiol Biochem 34. https://doi.
org/10.1159/000363026
205. Olas B, Wachowicz B (2002) Resveratrol and vitamin C as antioxidants in blood platelets.
Thromb Res 106. https://doi.org/10.1016/S0049-3848(02)00101-9
206. Tadolini B, Juliano C, Piu L, Franconi F, Cabrini L (2000) Resveratrol inhibition of lipid
peroxidation. Free Radic Res 33. https://doi.org/10.1080/10715760000300661
207. Ahmad KA, Clement MV, Pervaiz S (2003) Pro-oxidant activity of low doses of resveratrol
inhibits hydrogen peroxide – induced apoptosis. Proc Ann N Y Acad Sci 1010:365–373
208. Amato C (1994) Advantage of a micronized flavonoidic fraction (Daflon 500 mg) in compari-
son with a nonmicronized diosmin. Proc Angiol 45:531–536
209. Maksimović ZV, Maksimović M, Jadranin D, Kuzmanović I, Andonović O (2008)
Medicamentous treatment of chronic venous insufficiency using semisynthetic diosmin – a
prospective study. Acta Chir Iugosl 55. https://doi.org/10.2298/ACI0804053M
Steam Distillation: Principle
and Applications for the Extraction
of Essential Oils from Plants
Alankar Shrivastava
1 Introduction
There are more than 250 known essential oils. Various nations produce different
types of essential oils. India positions second on the planet for trading essential oils
[1]. The essential oil is volatile, and concentrated lipophilic liquid compounds with
characteristic fragrance are obtained from the various plants. These are also known
as fragrant oils, aromatic oils, volatile oils, or ethereal oils. They are also named on
the name of plant through which they are extracted, e.g., clove oil obtained cloves
[2]. Essential oil products cannot be replaced by synthetic materials. Most of these
are quite stable in normal atmospheric conditions. The fresh extractives are usually
colorless but may darken upon aging due to oxidation. This may be the reason why
they are kept in amber-colored glass bottles in dry place in properly closed contain-
ers. The industrial production of essential oil industry involves plant cultivation and
distillation to obtain oil [3]. It creates jobs for rural people for supply of raw materi-
als and workers involved in processing of oils. Essential oils are extensively used
globally for fragrances, food flavoring, pharmaceuticals, and aromatherapy [4].
The concentrated natural plant components containing volatile fragrant com-
pounds are essential oils (EOs), otherwise called volatile oils. Most EOs are inferred
by steam distillation and have remarkable properties, which bring about different
opportunities for application, likewise in the present medical context [5]. Essential
oils are secondary metabolites organic molecules and of relatively low molecular
weight rarely including sulfur, nitrogen, bromine, and chlorine in their structure [6].
The global essential oil market size was valued at USD 18.6 billion in 2020 and is
expected to expand at a compound annual growth rate (CAGR) of 7.4% in terms of
revenue from 2021 to 2028 [7]. By and large, medicinal oils are included in a few
significant parts in generally high concentration (20–95%) and other parts present
A. Shrivastava (*)
Shri Rawatpura Sarkar Institute of Pharmacy, Kumhari, Durg, India
in trace levels, e.g., Origanum compactum oil contains carvacrol (30%) and thymol
(27%), and citrus peel oil contains d-limonene (>80%). They are obtained from
various parts of the aromatic plants, including flowers, leaves, fruits, buds, seeds,
rhizomes, barks, and roots. Essential oils have recently begun to receive much atten-
tion as possible sources of safe and natural alternative medicines [8]. Essential oils
are known to possess anticancer, antioxidant, and antimicrobial effects. They may
act as an alternative for synthetic preservatives in food items. Gram-negative organ-
isms are believed to be slightly less sensitive to essential oils than Gram-positive
bacteria [9]. The components such as carvacrol and isoeugenol in some essential
oils have proven anti-inflammatory activity [10]. Essential oils in air-spray form can
be used for purifying indoor air and are often safe to allergic and asthmatic patients
[11]. Some essential oils, e.g., clove, lavender, and eucalyptus oils were also found
effective against many fungal species isolated from environmental air samples [12].
2 Distillation
Techniques for the purification of chemicals have a place within the domain of parti-
tion science. Every method for separation of one component from others can be
used for purifying components. The significant general thought for the improve-
ment of purity of any component is the selection of materials for the construction of
apparatus in which purification tasks are to be performed. Distillation is one of the
well-known techniques usually performed in the initial stages of purification. This
can be combined with other processes, e.g., chromatography, if high-grade purifica-
tion is required [13].
Distillation is a commonly used process for separation of liquid mixtures into
pure components and is an important unit operation for running chemical, petro-
chemical, and related industries. The normal perception is that it will remain an
important operation in these industries in the future as well. Advances in distillation
procedures have been basic to expanding the way of life for mankind in the course
of the past few thousand years [14]. Distillation is the most established separation
process and the most broadly involved unit operation in industry. The technique can
be utilized for many different purposes (Also refer Fig. 1) [15].
Innovations in the field of distillation processes can promote sustained growth in
food and chemical industries [16]. The term distillation alludes to an overall class of
strategies used to isolate parts from a blend dependent on a distinction in their vola-
tilities [17, 18]. By and large, a distillation process involves heating the liquid mix-
ture to the vapor state so as to enable the selective condensation and partition of
desired component(s) [19, 20]. The thermodynamic equilibrium governs the degree
of partition while the pace of division is controlled by the mass exchange [21, 22].
The basic principle of separation is presented in Fig. 2.
Steam Distillation: Principle and Applications for the Extraction of Essential Oils… 895
3 Brief History
Traditional extraction advances to recuperate esteem added items from plant materi-
als incorporate solvent extraction, steam distillation, and acid and alkali extraction.
In these, steam distillation method is known for recovery of high-boiling-point vola-
tile oils from complex and inert components, solid or liquid, using superheated or
saturated steam for separation [22]. Although it is unknown exactly what was dis-
tilled, the finding of clay jars from Babylonia that resemble stills may be the first
proof of distillation. The carbon dating studies show that those pots were of second
millennium BC. Albeit the principal drawings of stills date from the Hellenistic
school of chemists (second century AD), from composed and archeological proof it
was presumably the Arab chemists who idealized the craft of distillation to a degree
important to get genuinely focused alcohol from a fermented beverage. The Arabs
turned into the world’s driving researchers and were quick to involve distilled alco-
hol in medication. The soonest piece of distillation device (from Iran) that has
endured is Islamic and is dated between the tenth and twelfth hundreds of years. In
the nineteenth century, the alcohol business extended extensively, particularly in
France. The distillation of alcoholic beverages was becoming inescapable all
through Europe and Asia. The earliest stills of the eastern Asians and Arabs were
relatively small earthenware or glass pot stills of the alembic type, but did not use
flowing water as a coolant. The initial distillation assembly soju, which is an exam-
ple of Korean traditional stills, was heated by fires by charcoal or wood and recently
(in bygone eras) by boiling water or steam. Later copper replaced the material uti-
lized for making stills because of its advantages such as durability, good thermal
conductivity, and malleability. To some extent, bronze was also used for the same
purpose. Copper stills could be made to a wide scope of shapes and sizes; they could
persevere through quite a long while of utilization, didn’t break when warmed
straightforwardly, and caused a more uniform pace of distillation, with minimal
charring. The additional advantage of copper stills is improved organoleptic proper-
ties of distilled spirits due to removal of volatile organosulfur compounds [23].
Throughout the long term, the method spread broadly, and the principal book
regarding the matter, Das kleine Distillierbuch, by Brunswig, appeared in 1500
[24]. In science, the synthetic course of steam distillation was first portrayed by
Avicenna (Ibn Sina, 980–1037, Persia, Afghanistan). The procedure was utilized to
deliver essential oils and alcohol; the former was basic to fragrance-based treatment
(aromatherapy). Another invention was condensing coil increases the effectiveness
of distillation of essential oils using steam distillation [25].
Before the tenth century, it was still generally accepted that most essential oils
had solid therapeutic properties. Therefore, most of the credit of improvement in
purification and recovery of essential oils using distillation technologies goes to
pharmacists [26]. Jean-Baptiste Cellier-Blumenthal invented the first continuously
working distillation column in France and patented it in 1813. The first steam distil-
lation was probably performed by Claude Dariot (1533–1594), who heated the ves-
sel part between bottom and head. The direct steam distillation, however, was
Steam Distillation: Principle and Applications for the Extraction of Essential Oils… 897
reported by the Chinese around seventh century, but the evidence is not conclu-
sive [27].
3.1 Steam Distillation
Steam distillation facilitates the separation of compounds that are steam volatile
below their normal boiling points. The water obtained in the receiver can be easily
removed from volatile portion. One of the examples is the separation of naphthalene
and nitrobenzene [28]. This method is traditionally used for the separation of essen-
tial oils from plants. The superheated or saturated steam is responsible for vaporiza-
tion or extraction of fragrant compounds from the raw material. The volatile
components absorb heat from steam and are transported along with water vapors.
The components reached to the condenser and formed organic layer above water in
the receiver. Overall, two products are obtained in this process: (1) hydrosol and (2)
volatile oil. The hydrosol consists of water and some hydrolyzed components of
plant forming the bottom layer. The volatile oil in the upper layer in the decanter can
be then easily separated [29].
3.2 Principle
As per the kind of contact between the network and the water as well as steam, there
are three variations of the steam distillation process: dry steam distillation (Fig. 3),
direct steam distillation (Fig. 4), and hydrodistillation (water distillation) (Fig. 5).
distinctions are because of the steam being created external to the still and super-
heated at moderate pressures [30]. In comparison with other extraction processes,
steam distillation presents some advantages:
• Products obtained are solvent-free
• Further separation steps are not required
• Possibility of large-scale production
• Cheap equipment and well-known technology
• Minimum loss of polar compounds by controlling the reflux
However, the problem associated with steam distillation is degradation of ther-
mally unstable components and/or hydrolysis reaction with water, long extraction
period (1–5 h), high-power requirement, polluted waste water discharge, and risk of
foam or emulsion formation [31].
The blend of immiscible liquids tends to boil when the total vapor pressure is
equivalent to the atmospheric pressure. The principle of steam distillation can be
explained by example of distillation of mixture of turpentine and water. The boiling
point of turpentine is about 160 °C, but the mixture with water boils at about
95.6 °C. The vapor pressure of combination of turpentine and water reaches to
atmospheric pressure at lower temperature. The vapor pressures of individual com-
ponents at this point are about 15.06 kPa (113 mmHg) and 101.31 kPa for water and
turpentine, respectively. Thus, using this principle, essential oils with high boiling
point may also be separated easily with less deterioration and loss [32]. Furthermore,
countercurrent stream of vapour and liquid in a distillation column expands mass
exchange effectiveness. Superheated steam is likewise frequently utilized [33, 34].
Electromagnetic waves with frequency between 300 MHz and 300 GHz are called
microwaves. The combination of ionic conduction and dipole movement is respon-
sible for heating. The mechanism of working of microwave is out of scope of writ-
ing, readers may refer any literature (also refer Fig. 6) related to its working
mechanism. The advantages are better control, almost instantaneous heating and
less possibility of degradation of components. On the other hand, not all substances
can be heated using microwave and depends on the polarity of solvent, e.g., hexane
and chloroform (less polar) not produces heat. Overall, the efficiency of distillation
is increased by using microwave technology. There are many literatures published
based on this method. The solvent-free microwave-based distillation methods is
further extension [35].
In this process, distillation is carried out under normal pressure without solvent
by placing fresh leafy material in microwave reaction apparatus. The oil cells bursts
due to increase in inner tension when irradiated with microwaves and released oil
then distilled by moisture present in situ water from fresh leaves [36, 37]. A few
examples are extraction from Schisandra chinensis [38], Korean medicines [39],
orange peels [37], and Thymus mastichina [40].
900 A. Shrivastava
4 Future Aspects
Acknowledgments The authors acknowledge the support given by KIET Group of Institution,
KIET School of Pharmacy, Ghaziabad.
References
37.1 Introduction
Plants are the key to life on earth. They are, directly or indirectly, the primary source
of energy for all terrestrial animals; for instance, plants supply directly 90% of calo-
rific intake and 80% of the protein intake of man. Breeding of crop plants has been
carried out by man for thousands of years. It is, however, only over the last 50 years,
as a result of highly sophisticated breeding processes, combined with improved agri-
cultural methods and modern technology, that this has brought about a dramatic
increase in yield and quality of crops. Generally, though, these improved crop plants
are often susceptible to many diseases caused by fungi, insects, bacteria, nematodes,
and viruses. The tendency for crop plants to be threatened by many diseases and pests
compared to wild plant species is due mostly to the breeding programs, whereby
selection for characteristics such as yield takes priority over those for disease and
pest resistance. For many years this has been overcome by the use of pesticides, but
there is now increasing concern about the environmental safety of these chemicals,
which can persist in the food chain and may be toxic to plants and animals. Given that
pesticides have been largely successful only in the control of fungi and insects, and
offer little protection against viruses, viroids, and bacteria, it is now more urgent than
ever to find alternative methods of protecting crop plants from diseases [1].
Plant breeding has several other serious limitations for use as a tool to increase
disease resistance. There are only a limited number of plant species which are able
to cross-fertilize, thus restricting the transfer of potentially useful traits. Moreover,
having found useful traits, it is impossible to prevent the co-transfer of undesirable
ones, which can take many years to breed out again by back-crossing.
How can genetic engineering be of use in the quest for new answers to the prob-
lems of providing plants with the ability to resist disease? The aim of crop plant
genetic engineering is to insert a gene (or genes) which improves an existing plant
variety whilst retaining the desirable genetic make-up of the original plant. The
main tool at the disposal of the scientist is the use of nature’s own genetic engineer,
Agrobacterium. Three different species of Agrobacterium are found, i.e., A. tumefa-
ciens, A. rhizogenes, and A. radiobacter of which A. radiobacter is an avirulent
strain and A. tumefaciens is found to be most effective.
The microbe Agrobacterium tumefaciens is harmful to plants and useful to scien-
tists for the same reason: It transfers DNA into plant genomes. Found in soil world-
wide, A. tumefaciens causes disease in plants by transferring its own DNA into plant
cells. But in the laboratory, the ability to move all sorts of genes into plants has
made the microbe the standard tool for investigating plant genetics and modifying
crops. The manipulation of this bacterium’s natural functions has allowed the biolo-
gist to transfer many foreign genes into plants.
infects a plant, the bacterium travels throughout the root system and can wipe out an
entire crop. The only option for farmers is to destroy the plants. The practice of
replacing the tumor-inducing genes with other DNA began in the 1970s and led to
the widespread use of the bacterium in research. The microbe is particularly well
suited for testing the functions of individual plant genes because bacterial DNA
disrupts the genome at the point of insertion.
“Agrobacterium can be used to insert a piece of DNA in the middle of a plant
gene, thus inactivating the gene,” says Derek W. Wood, who led part of the University
of Washington project. “We can then analyze the mutation to see what it does to the
plant.” This experiment has been done on a large scale. Many thousands of mutant
potato and alfalfa plants are available to plant researchers from institutions like the
University of Wisconsin in Madison. The microbe has also been used to create
transgenic crops, including new strains of corn and soybeans. Agrobacterium-
mediated gene transfer into a wide variety of crop species has already become a
realizable technique for plant improvement by transferring agronomically useful
genes such as insect resistance [2], virus resistance [3], resistance against bacterium
[4], and herbicide resistance [5, 6].
Agrobacterium tumefaciens genome has a very unusual structure. Some 5400
genes reside on four DNA elements—a circular chromosome, a linear chromosome,
and two smaller circular structures called plasmids. Many bacteria have circular
chromosomes and some have linear chromosomes, but Agrobacteria are the only
species known to have both structures together.
The discovery of the importance of the Ti plasmid in crown gall raised the next
obvious question, what is its role in tumor formation? The stable properties of tumor
tissue suggested that the plasmid, or some part of it, was stably maintained in the
plant cell. Thus, it became critical to demonstrate that plasmid DNA was, in fact,
actually present in tumor tissue. The Seattle group had previously searched for
908 D. P. Barik
bacterial DNA and then for the entire Ti plasmid in tumor tissue without success. At
this time, Southern hybridization was not yet developed. The laborious technique
they used was DNA renaturation kinetic analysis in which the rate of association of
a small amount of 32P-labeled single-stranded DNA (probe DNA) into a double-
stranded form is measured in the presence of a large amount of either tumor or
normal plant tissue DNA (driver DNA). If the tumor DNA contained DNA homolo-
gous to the probe, the tumor DNA would accelerate the association of the single-
stranded probe DNA. However, the drawback of this method is that it can detect
foreign DNA only if it constitutes a significant fraction of the labeled probe. Thus,
if only a few genes of the megaplasmid were present per tumor cell, they would not
be detected. Since the Seattle group did not detect the entire plasmid, they were bet-
ting that only a small part was in tumor cells. They mounted a brute force effort to
demonstrate this. Since it was likely that tumor tissue contained both transformed
and untransformed cells, as a first step, Milt Gordon went to George Melcher’s lab
in Germany where he cloned a tumor line. The renaturation kinetic analysis became
an assembly line operation, with different members of the lab playing specific roles.
As soon as the 32P-d CTP arrived, a nick translation reaction was initiated to label
the plasmid to its maximum specific activity. The labeled plasmid was then digested
with the restriction enzyme SmaI, which we had to purify since it was not commer-
cially available. The fragments were separated by preparative gel electrophoresis
and the 15 resolvable bands were electroeluted from the gel slices. Seventy-five
renaturation kinetic assays were set up with labeled probes and denatured driver
DNAs from either tumor or normal tissue. A total of 525 samples were taken around
the clock, and the percentage of single-stranded probe that had renatured to double-
stranded DNA at each time point was determined. These experiments gave the
hoped-for result. Two labeled bands renatured faster when incubated with tumor,
but not normal plant DNA. No other bands did. Thus, we concluded that a fragment
of the Ti plasmid was indeed present in tumor cells. The suspicions of many inves-
tigators had proven correct! DNA, and more specifically a fragment of the Ti plas-
mid, now named T-DNA for transferred-DNA, was the long-sought-after TIP.
Mike Thomashow in Seattle analyzed four cloned tumor lines, using restriction
enzymes and molecular hybridization techniques, to determine how T-DNA is
maintained in the host cell genome. Fortunately, by this time Southern hybridization
was a proven and reliable technique. Following digestion of tumor DNA with
restriction enzymes that cleaved within or outside the T-DNA and by determining
the sizes and numbers of the resulting T-DNA molecules by Southern hybridization,
he concluded (1) that the T-DNA is integrated into plant DNA, (2) that preferred
regions of the Ti plasmid serve as points of attachment to the plant DNA, and (3)
that the T-DNA can be linked to more than one site in plant DNA. Later studies by
Narendra Yadav in Mary-Dell Chilton’s group demonstrated that T-DNA is flanked
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants... 909
by 25-bp direct repeats. Presumably any DNA bounded by these sequences would
be transferred to the plant. Studies from several laboratories using Southern hybrid-
ization on fractionated cell components demonstrated that T-DNA is integrated only
into nuclear DNA.
The question of whether the integrated T-DNA is transcribed was quickly
answered by Martin Drummond working with Mary-Dell Chilton. They demon-
strated that RNA isolated from a cloned tumor line labeled with 32P in vivo hybrid-
ized to T-DNA. Interestingly, T-DNA is not transcribed in Agrobacterium.
Although the T-DNA with its two flanking border sequences had now been identi-
fied, how it induced crown gall tumors and what other genes are important in plant
cell transformation awaited discovery. The Ghent, Leiden, and Seattle groups were
the primary contributors to this part of the story. In Ghent, Marcel Holsters led the
effort to determine the functional organization of the Ti plasmid of strain C58.
Using transposons Tn1 and Tn7, she isolated insertion and deletion mutants and
described their oncogenic and other biological properties. These studies, as well as
those from the two other groups, revealed that another region of the Ti plasmid
besides the T-DNA is essential for oncogenesis. Insertion of transposons in this
region, now termed the virulence or vir region, created an avirulent phenotype. They
also identified regions in the T-DNA and elsewhere on the plasmid that are not
essential for oncogenicity.
By mutagenizing the entire genome, Dave Garfinkel in Seattle uncovered a num-
ber of loci in the chromosome that are also important for virulence. They included
loci-encoding proteins required for binding of Agrobacterium to plant cells, a sugar-
binding protein required for maximum vir gene induction, and loci required for
sensing the acidic environment required for vir gene induction. Garfinkel also used
site-directed mutagenesis with Tn5 to develop a fine structure map of the T-DNA in
order to identify the function of genes responsible for tumor formation. He, together
with several colleagues in Seattle and Gert Ooms, a visiting graduate student from
Rob Schilperoort’s lab, isolated 75 Tn5 and 3 Tn3 insertions which identified four
genetic loci. Tms mutants (tumor morphology shooty) resulted in shoot prolifera-
tion in the tumor; tmr (tumor morphology rooty) resulted in root formation; and
insertions in tml (tumor morphology large) gave rise to unusually large tumors on
certain plants. Gert Ooms made similar observations independently. Later studies
showed that the “shooty” mutations resulted from insertions in two loci (iaaH and
iaaM) concerned with auxin synthesis and the “rooty” mutations contained an inser-
tion in a gene for cytokinin synthesis (ipt). Interestingly, no single insertion resulted
in avirulence. In later studies, Walt Ream showed that an avirulent phenotype
required simultaneous insertions in both genes coding for auxin and cytokinin
biosynthesis.
910 D. P. Barik
The process of transfer of T-DNA from Agrobacterium to the cells of the host plant
can be divided into a bacterial step and a plant cell step [7]. Once T-DNA was identi-
fied and characterized, attention turned to the question of how Agrobacterium trans-
fers T-DNA into plant cells. The vir region of the Ti plasmid is highly conserved in
octopine and nopaline strains and was surmised to be involved in the transfer pro-
cess. As a first step to constructing a fine structure map of this region, Scott Stachel
and Gyn An in Seattle constructed a Tn3 lacZ transposon which could randomly
generate gene fusions as well as serve as a reporter to study gene expression. They
identified six complementation groups in this region. In the course of these studies,
Stachel also observed that expression of the vir genes was enhanced significantly by
exudates of a variety of plant cell cultures. From Seattle, Stachel went to Ghent
where he and his collaborators then identified two phenolic compounds, acetosyrin-
gone (AS) and hydroxyacetosyringone (OH-AS) in plant exudates that induce the
vir genes [8].
Stachel was also an important player in elucidating the mechanism by which AS
induces the vir genes. He and Steve Winans in Seattle independently characterized
a two-component system, VirA/G, which controls expression of the vir regulon. AS
binds to the membrane-bound histidine kinase sensor protein VirA, which then acti-
vates the response regulator VirG, which in turn binds to upstream regions of each
of the vir operons and activates their transcription. This particular two-component
regulatory system was one of the very first to be described in any organism.
Stachel and Pat Zambryski also demonstrated that the addition of AS to
Agrobacterium results in the formation of a single-stranded, linear DNA molecule,
which they suggested is the intermediate in the transfer of T-DNA to plant cells.
They further suggested that this form of DNA, now termed the T-strand, resulted
from an endonucleolytic cleavage at the right and left borders of the T-DNA and
compared the transfer of this DNA to the transfer of single-stranded DNA in bacte-
rial conjugation.
The publication of Stachel et al. was quickly followed by a paper by Martin
Yanofsky et al. in Seattle which demonstrated that the virD operon encodes a site-
specific endonuclease that cleaves within each of the 25-bp direct repeats that flank
the T-DNA. Two proteins are required, Vir D1 and Vir D2. Once the Vir D2 protein
cleaves the DNA it remains covalently attached to the 5′ end of the T-strand, as first
demonstrated by Calvin Young et al. This protein likely serves as a pilot protein for
entry of the T-DNA into the plant cell nucleus. In support of this idea, Luis Herrera-
Estrella in Ghent demonstrated that the amino-terminal portion of the Vir D2 pro-
tein contains a nuclear localization signal. Another Vir protein Vir E2 was shown by
Barbara Hohn et al. in Basel, Switzerland, to be a single-stranded DNA-binding
protein which also has nuclear localization signals. This protein not only protects
the T-strand from degradation by plant nucleases but also helps direct the T-strand
into the plant cell nucleus (Fig. 2).
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants... 911
Fig. 4 A case study using Agrobacterium tumefaciens in legume plant, i.e., Lathyrus sativus L.
known. Once the T-DNA is transferred into the plant, the vir regulon no longer
needs to be expressed. Therefore, one of the products of T-DNA expression, indole
acetic acid (IAA), shuts down expression of the vir regulon by inhibiting the activa-
tion of the VirA/G regulatory system, thereby conserving carbon and energy. At
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants... 915
slightly higher concentrations, IAA kills the bacteria which may help explain the
long-standing observation that crown gall tumors often do not contain viable
bacteria.
Another product of transformed plant cells which plays an important role in the
biology of Agrobacterium are the opines. Studies from the laboratories of Stephen
Farrand, Steve Winans, Allen Kerr, and Max Tate demonstrated that certain opines
induce the synthesis of a signal molecule acyl homoserine lactone, which interacts
with a transcriptional activator to promote conjugal transfer of the Ti plasmid to
other agrobacteria.
Not only does Agrobacterium utilize extracellular plant signals in the infection
process, but it also hijacks cellular processes for the transport of the T-strand to the
nucleus. These pioneering studies have been carried out largely in the laboratories of
Stanton Gelvin, Vitaly Citovsky, and Tsvi Tzfira. The viscous cytoplasm with its
assemblage of membranous and nonmembranous structures most likely requires that
the T-strand be actively transported to the nucleus. Many DNA viruses use dynein
motor proteins of the host and the host microtubule system for their transport to the
nucleus. The available data suggest this may be true for the journey of the T-strand
to the nucleus also. Both Vir D2 and Vir E2 proteins with their nuclear localization
signals are important in nuclear import and both accumulate in the plant cell nucleus.
A number of other host proteins have been implicated in their interaction with the
T-strand but the roles they play in the transformation process are obscure.
Soon after it was shown that T-DNA was integrated into the plant cell nucleus and
that the T-DNA was defined by its flanking border sequences, many laboratories
realized that Agrobacterium potentially might be a very useful vector for introduc-
ing any desired DNA into plants. This possibility was strengthened when Léon
Otten in Jeff Schell’s group demonstrated that opine genes were transferred in a
Mendelian fashion. Self-pollination of the transformed plant and crossing of trans-
formed and untransformed plants showed that this trait was transmitted through
pollen and eggs as a single dominant gene with the predicted Mendelian ratios.
Jean-Pierre Hernalsteens then inserted a Tn 7 transposon into an opine locus and
demonstrated that it was transferred to, and maintained in, tumor tissues induced by
this mutant strain. Since Tn5 had its own promoter, antibiotic resistance was not
expressed. However, this experiment did demonstrate that using the Ti plasmid as a
vector system for introducing DNA into plant cells was feasible.
The major technical problem in using the Ti plasmid as a gene vector is its large
size. This makes it impossible to employ a direct recombination strategy to insert
genes of interest into the T-DNA. Several indirect methods were developed in the
early 1980s for manipulating T-DNA but these were cumbersome and not easily
carried out. The problem was solved when two independent groups, one led by
Mary-Dell Chilton and the other by Rob Schilperoort, shrank the plasmid by
916 D. P. Barik
separating its two essential regions into two plasmids; the 25-bp borders flanking
the T-DNA between which genes of interest could be inserted and the vir region
which is necessary for the processing and transfer of the T-DNA. This binary vector
system is used in most laboratories today.
Two additional hurdles remained before plants could be routinely transformed in
an industrial setting with genes that create added value on the engineered plants.
First, plants had to be regenerated with the genes of interest stably maintained and
second, the introduced genes had to be expressed. The first problem was solved by
two independent groups Pat Zambryski et al. in Jeff Schell’s group and Andy Binns
et al. at the University of Pennsylvania. They both showed that the oncogenes of
T-DNA interfere with normal plant cell differentiation and that if they are removed,
transformed cells readily regenerate into normal plants. The second hurdle was
overcome when three independent groups (the Ghent group, a group from Monsanto
headed by Rob Fraley, and Chilton’s group) almost simultaneously reported on the
expression of foreign genes in transformed plants. All three groups used the nopal-
ine synthase promoter fused to an antibiotic resistance locus which resulted in anti-
biotic resistant tobacco transformants. These experiments opened up the age of
plant genetic engineering with the Monsanto Company leading the subsequent
industrial revolution in agriculture.
Although Agrobacterium has a very broad host range and tumors have been
detected on many dicotyledonous plants in Nature, the most important plants eco-
nomically worldwide are the monocots, which include the cereals. Since crown gall
tumors had never been observed in this group of plants, and initial efforts to trans-
form many of them in the laboratory were unsuccessful, many agrobiologists
believed that monocots were totally recalcitrant to Agrobacterium transformation.
However, a clever experiment performed in Barbara Hohn’s group, on the sugges-
tion of her virologist husband, clearly demonstrated in fact that Agrobacterium was
able to carry out the initial steps in maize transformation—the transfer of T-DNA
and expression of the transferred genes. These investigators introduced tandemly
repeated copies of the genome of maize streak virus between the T-DNA borders
and inoculated maize plants with Agrobacterium carrying this construct in a binary
vector. The inoculated leaves developed symptoms of maize streak viral disease!
Further experiments demonstrated that this viral infection, termed agroinfection,
requires the same Vir proteins as are required for tumor formation by wild-type
Agrobacterium. These results encouraged others to test a variety of strains of
Agrobacterium and different monocot tissues using a variety of reporter systems
and selective markers to look for T-DNA transfer and expression. In 1994, Hei and
his colleagues at the Japan Tobacco Company reported the efficient transformation
of rice. They studied a variety of tissues and several different strains of Agrobacterium,
screening for GUS color and selecting for hygromycin resistance. Of all the tissues
tested, scutellum callus gave the highest level of transformation and a “super-
binary” vector gave especially high transformation frequencies of various cultivars
of Japonica rice. High-frequency Agrobacterium transformation has now been
achieved for most cereal crops.
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants... 917
Although Agrobacterium gene transfer and crown gall tumor formation in a wide
variety of plants is a common occurrence in nature, in the laboratory, Agrobacterium
can transfer DNA into a much broader group of eukaryotic cells. This was first dem-
onstrated by Paul Hooykaas and colleagues who reported that Agrobacterium carry-
ing a ura+ locus between T-DNA borders could transform a ura– strain of the yeast
Saccharomyces cerevisiae to prototrophy in medium that promoted vir gene induc-
tion. This group also showed that many other fungi could be transformed following
co-cultivation with Agrobacterium. Now, numerous algae, protozoa, and even Hela
cells have been stably transformed in the laboratory. These reports have opened up
a whole new use for Agrobacterium, allowing the genetic analysis of organisms that
previously were totally recalcitrant to such studies (Tables 1 and 2). A standard
protocol for Agrobacterium-mediated transformation of Lathyrus sativus is given in
Figs. 3 and 4.
Agrobacterium is truly a natural genetic engineer, in that not only does it transform
plants to synthesize food (opines) for itself but its DNA can be found even in per-
fectly normal appearing, uninfected plants. In Seattle, Frank White and colleagues
reported that the T-DNA (rol) genes of A. rhizogenes can be readily detected in
uninfected Nicotiana glauca and many other species of tobacco. It seems most
likely that A. rhizogenes infected a Nicotiana plant millions of years ago to form
hairy root tumors which then regenerated into normal appearing plants which passed
down the conserved sequences to their progeny. This observation puts to rest the
idea that transgenic plants represent an unnatural phenomenon that only occurs in
the laboratory. One can safely bet that the finding of T-DNA in many species of
Nicotiana is not an isolated phenomenon limited to this plant.
Research on crown gall tumors began as a classic study in plant pathology aimed at
identifying the cause of a devastating plant disease with unusual symptoms. Studies
on Agrobacterium by investigators in laboratories around the world have revealed
new biological phenomena and practical applications far beyond what Smith and
Townsend could ever have imagined. On the one hand, Agrobacterium causes seri-
ous problems for growers of grapes, stone fruits, and ornamental plants. On the
other hand, it has provided an unusually exciting and rewarding experimental sys-
tem for those of us fortunate to have entered into its study. The study of
Table 1 Genetic transformation of tree species using Agrobacterium tumefaciens
918
Kanamycin
Infection and conc. for
co-cultivation selection (μg/ Gene
Species Strain Construct Explant details ml) expression Detection method References
Actinidia deliciosa LBA4404 pRokla-EG Leaf disc Infected with 50 nptII, Soybean PCR and Southern blot [9]
(kiwi fruit) nptII, soybean petiole, stem bacteria β-1,3 analysis, Northern blot
β-1,3 segment endonuclease and Western blot
endonuclease analysis
Carica papaya LBA4404 pB1121 (nptII, Petioles Just dipped and 50 gus, gus PCR and Southern blot [10]
(papaya) gus) from co-cultivated for analysis, Northern blot
multishoot 2 days and Western blot
analysis
Citrus aurantifolia EHA 105 p35SGUSINT Internodal Infected with 10 nptII, gus, npt, PRC and Southern blot [11]
(lime orange) (nptII, gus) segment bacterium for gus analysis, ELISA and
15–30 mins and histochemical enzyme
then assay
co-cultivated for
3 days
Citrus aurantium EHA 101 pGA482GG- Internode Explants 100 ctv-cp, gus, PCR and Southern bot [12]
(sour orange) CTVCP (gus, co-cultivated for ctv-cp analysis, Histochemical
ctv-cp) 1–3 days enzyme assay, Western
blot analysis
Citrus sinensis – EHA 105 p35SGUSINT Epicotyl Explants 100 nptII, gus, gus PCR and Southern blot [13]
Osbeck × (nptII, gus) incubated 15 min analysis, Northern blot
Poncirus trifolia in 15 ml bacterial and Western blot
(citrange) suspension, analysis, Histochemical
co-cultivated for enzyme assay
1, 3, 5 days
D. P. Barik
Kanamycin
Infection and conc. for
co-cultivation selection (μg/ Gene
Species Strain Construct Explant details ml) expression Detection method References
Citrus sinensis LBA4404, p35SGUSINT Epicotyl Epicotyl 100 nptII, gus, gus PCR and Southern blot [14]
(Washington novel EHA 101, (nptII, gus) segments analysis, Northern blot
orange) EHA 105, C58 infected for and Western blot
10 min in analysis, histochemical
bacterial enzyme assay
suspension
(5 × 108 cfu/ml)
blotted dry,
co-cultivated for
2–3 days
Diospyros kaki LBA 4404, pB1121, Hypocotyl Segments of 100 nptII, gus, gus PCR and Southern blot [9]
(Japanese EHA 101 pSMAK251 hypocotyl analysis, Northern blot
persimmon) (nptII, gus) immersed in and Western blot
bacterial analysis, histochemical
suspension enzyme assay
approximately
(5 × 108 cfu/ml)
for 15 min and
then
co-cultivated for
3 days
Eucalyptus A6, LBA Each Leaf disc Co-cultivated for 9 nptII, gus, gus Southern blot analysis, [15]
camaldulensis 4404, GV containing petiole, stem 2 days in NPT II enzyme assay,
(eucalyptus) 3111, AGL1, nptII, gus segment bacterial histochemical enzyme
GV3850 suspension (109 assay
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants...
cells/ml) on
callusing
medium
919
(continued)
Table 1 (continued)
920
Kanamycin
Infection and conc. for
co-cultivation selection (μg/ Gene
Species Strain Construct Explant details ml) expression Detection method References
Hevea brasiliensis GV 2260 p35SGUSINT Callus Calli submerged 100 nptII, gus, Southern blot analysis, [16]
(rubber) (nptII, gus) in an overnight nptII, gus, gus histochemical enzyme
grown assay by Sudan III
Agrobacterium Staining (Jensen 1962)
suspension for
1 min and then
co-cultivated for
2 days
Larix C58 pPMP90, Somatic Bacterial 50 nptII PCR and Southern blot [17]
kaempferi × L. pMRKE70Km embryo suspension at OD analysis
decidua (hybrid (2 copies of 0.3
larix) nptII) (approximately
108 cfu/ml)
applied to
embryonal
masses kept on
proliferation
medium in dark
for 48 h
Malus domestica EHA 105 p35SGUSINT Internode Epicotyl 100 gus, gus Histochemical enzyme [18]
(royal gala apple) (nptII, gus) segments dipped assay
in overnight
grown bacterial
suspension
culture, a density
of 2 × 108 cfu/ml,
blotted dry,
D. P. Barik
co-cultivated for
48 h
Kanamycin
Infection and conc. for
co-cultivation selection (μg/ Gene
Species Strain Construct Explant details ml) expression Detection method References
Malus domestica EHA 105 p35SGUSINT Leaf disc Wounded leaves 25 nptII, gus, PCR and Southern blot [19]
(Marshal (nptII, gus) petiole, stem co-cultivated in nptII, gus, gus analysis, ELISA and
Maclntosh apple) segment dark for 48 h NPT enzyme assay by
after infecting histochemical enzyme
with bacterial assay
suspension
Populusalba×grand- C58 pPMG85/587 Leaf 1–5 ml bacterial 50 nptII, aro A, Southern blot analysis
identata (populus (nptII, EPG segment broth culture nptII, aro A
hybrid NG 5339) synthase, aro infected for
A) 30 min and then
co-cultivated for
48–96 h
Prunus dulcis LBA 4404, p35SGUSINT Leaf Leaf wounded 15 nptII, gus, gus Southern blot analysis, [20]
(almond) EHA 101, (nptII, gus) from mid-rib NPT II enzyme assay
EHA 105, C58 dipped in and ELISA,
bacterial histochemical enzyme
suspension and assay
co-cultivated for
3–4 days
Pyrus communis EHA 101 pFM3002, Leaf Infected with 100 nptII, uid A, PCR and Southern blot [21]
(pear) pFAJ300 bacterial attacin E, analysis, Northern blot
(nptII, uid A, suspension attacin E and Western blot
attacin E) 5 × 107 cuf/ml analysis
Santalum album LBA4404, pKIW 105, Somatic Infected with 50 nptII, uid A, PCR and Southern blot [22]
(sandalwood) EHA 101 pIG121-Hm embryo bacterial nptII, uid A analysis, NPT II
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants...
Kanamycin
Infection and selection
Species Strain Construct Explant co-cultivation details (μgml−1) Gene detection and expression References
Cajanus cajan L. and LBA4404 pCAMBIA 1303 Different Infection for 30 min 50 uid A Histochemical GUS [23]
some grain legumes (uidA and gus) explants and co-cultivation for gus assay
48 h
Cajanus cajan (L.). GV2260 CaMV 35S Embryonic axis Co-cultivation for 12 h 50 nptII PCR analysis [24]
Millsp. (nptII) Southern analysis
Vr. Pusa 885
Cajanus cajan L. EHA105 pBin9GusInt Cotyledonary Infection for 20 min 50 nptII GUS-histochemical [25]
Millsp. (nptII and gusA) node and leaf and co-cultivation for gusA assay
explants 20 h
Cicer arietinum L. LBA4404 pBI 121 Embryo axis Infection for 20 min 50 nptII Histochemical GUS [26]
CaMV35S Co-cultivation for gus assay
(nptII and gus) 2–3 weeks Southern analysis
Cicer arietinum L. LBA4404 pBI 121 Embryo axis Cultured for 24 h on 50 nptII NPT II enzyme assay [27]
genotypes (uidA and nptII) SR medium prior to gus Histochemical GUS
ICCV-1 co-cultivation with assay
ICCV-6 bacteria Southern analysis
and Desi (local)
Cicer arietinum L. vr. A281 pKIW1105 Stem Co-cultivation for 80 nptII Histochemical GUS [28]
6153 and CM72 C58 p35SGUSINT Leaf 2 days gus assay
(nptII and gus)
Dalbergia sissoo EHA105 p35SGUSINT Cotyledonary Infection for 8–15 min 50 nptII GUS-histochemical [29]
(nptII and gus) node Co-cultivation for gus assay
Epicotyl 2–4 days
Galega orientalis Lam. LBA4404 pAC 4404 Node Co-cultivation for 100 nptII GUS-histochemical [30]
AGL1 (nptII and gus) Cotyledon 4 days gus assay
EHA105 pTiBo 542
MOG301 pEHA 105
pMOG 301
CaMv35S and gus
Glycine max EHA105 pTIT37SE Cotyledon NAI 50 nptII gus Histochemical GUS [31]
D. P. Barik
Agropine type
pTiBo 542
Vigna unguiculata L. C58 pGV2260 Mature embryo NAI NAI gus GUS and DNA dot [42]
923
References
1. Old RW, Primrose SB (1994) Gene transfer to plants. In: Carr NG (ed) Principles of gene
manipulation, an introduction to genetic engineering. Blackwell Scientific Publications,
Oxford, pp 268–301. Tinland B (1996) The integration of T-DNA into plant genomes. Trends
Plant Sci 1:178–184
2. Ananda KP, Sharma RP, Malik VS (1996) The insecticidal proteins of Bacillus thuringiensis.
Adv Appl Microbiol 41:1–43
3. Beachy RN, Loesch-Fries S, Tumer NE (1990) Coat protein-mediated resistance against virus
infection. Ann Rev Phytopathol 28:451–474
4. Diiring K, Porsch P, Flaudung M, Lorz H (1993) Transgenic potato plants resistant to the phy-
topathogenic bacterium Erwinia corotovora. Plant J 3:587–598
5. Mohapatra U, McCabe MS, Power JB, Schepers F, Vander Arend A, Davey MR (1999)
Expression of the bar gene confers herbicide resistance in transgenic lettuce. Transgenic
Res 8:33–44
6. Mohapatra U, McCabe MS, Power JB (2002) Agrobacterium-mediated transformation of let-
tuce for bialaphus resistance. In: Nandi SK, LMS P, Kumar A (eds) Proceedings of the national
seminar on role of plant tissue culture in bio-diversity conservation and economic develop-
ment. Gynodaya Prakashan, Nainital, pp 445–455.s
7. Tinland B (1996) The integration of T-DNA into plant genomes. Elsevier 1(6):178–184
8. Mondal TK, Bhattacharya A, Ahuja PS, Chand PK (2001) Transgenic tea [Camellia sinensis
(L.) O Kuntze cv. Kangrajat] plants obtained by Agrobacterium-mediated transformation of
somatic embryos. Plant Cell Rep 20:712–720
9. Nakamura Y, Sawada H, Kobayashi S, Nakajima I, Yoshikawa M (1999) Expression of soya-
bean M, ß-endogluconase cDNA and effect on disease tolerance in Kiwifruit Plants. Plant Cell
Reports 18:527–532
10. Yang J-S, Yu T-A, Cheng Y-H, Yeh S-D (1996) Transgenic Papaya plants from Agrobacterium-
mediated transformation of petioles of in vitro propagated multi-shoots. Plant Cell Reports
15:459–464
11. Pena L, Cervera M, Juarez J, Navarro A, Pina JA, Navarro L (1997) Genetic transformation
of lime (Citrus aurantifolia Swing.): factors affecting transformation and regeneration. Plant
Cell Reports 16:731–737
12. Gutierrez-E MA, Luth D, Moore GA (1997) Factors affecting Agrobacterium-mediated trans-
formation in citrus and production of sour orange (Citrus aurantium L) plants expressing the
coat protein gene of citrus tristeza virus. Plant Cell Reports 16:745–753
Key Factors Influencing Agrobacterium-Mediated Transformation Efficiency in Plants... 925
13. Cervera M, Pina JA, Juarez J, Navarro L, Pena L (1998) Agrobacterium – mediated trans-
formation of citrange: factors affecting transformation and regeneration. Plant Cell Reports
18:271–278
14. Bond JE, Roose ML (1998) Agrobacterium-mediated transformation of commercially impor-
tant citrus cultivar Washington novel orange. Plant Cell Reports 18:229–234
15. Mullins KV, Llewellyn DJ, Hartney VJ, Strauss S, Dennis ES (1997) Regeneration and trans-
formation of Eucalyptus camaldulensis. Plant Cell Reports 16:787–791
16. Arokiaraj P, Yeang HY, Cheong KF, Hamzah S, Jones H, Coomber S, Charlwood BV (1998)
CaMV 355 promoter directs ß glucuronidase expression in the laticiferous system of trans-
genic Hevea brasiliensis (rubber tree). Plant Cell Reports 17:621–625
17. Leeve V, Lelu MA, Jouanin L, Cornu D, Pilate G (1997) Agrobacterium tumifaciens – medi-
ated transformation of hybrid Larch (Larix Kaempferi × L. decidua) and transgenic plant
regeneration. Plant Cell Reports 16:680–685
18. Liu Q, Salih S, Hummerschlag F (1998) Etiolation of ‘Royal Gola’ apple (Malus × domestica
Borkh.) shoots promotes high-frequency shoot organogenesis and enhanced ß-glucuronidase
expression from stem internodes. Plant Cell Reports 18:32–36
19. Bolar JP, Norelli JL, Harman GE, Brown SK, Aldwinckle HS (1999) Expression of fungal
chitinolytic enzymes in transgenic apples confers high levels of resistance to scab. Plant bio-
technology and in vitro biology 36:465–468
20. Miguiel CM, Oliveira C (1999) Transgenic Almond (Prunus dulcis Mill.) plants obtained by
Agrobacteriun-mediated transformation of leaf explants. Plant Cell Rep 18:387–393
21. Reynoird JP, Mourgues F, Norelli J, Aldwinckle HS, Brisset MN, Chevreu E (1999) First
evidence for improved resistance to fire blight in transgenic pear expressing the attacin E gene
from Hyalophora cecropia. Plant Sci 149(1):23–31
22. Shiri V, Rao KS (1998) Introduction and expression of marker genes in sandalwood (Santalum
album L.) following Agrobacterium mediated transformation. Plant Sci 131(1):53–63
23. Shrivastava DK, Sanyal I, Singh BD, Amla DV (2001) Endogenous GUS-activities in Cajanus
cajan L. and some grain legumes: selective suppression for expression of GUS reporter gene.
J Plant Biol 28(3):243–250
24. Lawrence PK, Koundal KR (2000) Simple protocol for Agrobacterium tumefaciens- mediated
transformation of pigeonpea [Cajanus cajan (L.) Millsp.]. J Plant Biol 27(3):299–302
25. Singh ND, Sahoo L, Sonia, Jaireal PK (2002) In vitro shoot organogenesis and plant regen-
eration from cotyledonary node and leaf explants of pigeon pea (Cajanus cajan L. Millsp.).
Physiol Mol Biol Plants 8(1):113–140
26. Fontana GS, Santini L, Caretto S, Frugis G, Mariotti D (1993) Genetic transformation in the
grain legume Cicer arietinum L. (Chickpea). Plant Cell Rep 12:194–198
27. Kar S, Johnson TM, Nayak P, Sen SK (1996) Efficient transgenic plant regeneration through
Agrobacterium-mediated transformation of chickpea (Cicer arietinun L.). Plant Cell Rep
16:32–37
28. Husnain T, Malik T, Riazuddian S, Gordon MP (1997) Studies on expression of marker genes
in chickpea. Plant Cell Tiss Org Cult 49:7–16
29. Pradhan C (2000) Plant regeneration from tissue & cell cultures and Agrobacterium-mediated
genetic transformation of Dalbergia species. Ph.D thesis, Utkal University, Vanivihar,
Bhubaneswar
30. Collen AMC, Jarl CI (1999) Comparison of different methods for plant regeneration and trans-
formation of the legume Galega orientalis Lam. (goat’s rue). Plant Cell Rep 19:13–19
31. Hinchee MAW, Connor-Ward D, Christine AN, McDonald RE, Sato SJ, Gasser CS, Fischott
DA, Re DB, Fraley RT, Horsch RB (1998) Production of transgenic soybean plants using
Agrobacterium-mediated DNA transfer. Bio/Technology 6:912–922
32. Meurer CA, Dinkins RD, Collins GB (1998) Factors affecting soybean cotyledonary node
transformation. Plant Cell Rep 18:180–186
33. Santarem ER, Trick HN, Essig JS, Finer JJ (1998) Sonicated-assisted Agrobacterium-mediated
transformation of soybean immature cotyledons: optimization of transient expression. Plant
Cell Rep. 17:752–759
926 D. P. Barik
34. Zhang Z, Xing A, Staswick P, Clemente TE (1999) The use of glufosinate as a selective agent
in Agrobacterium mediated transformation of soyabean. Plant Cell Tiss & Org Cult 56:37–46
35. Barna KS, Mehta SL (1995) Genetic transformation and somatic embryogenesis in Lathyrus
sativus. J Plant Biochem Biotechnol 4:67–71
36. Ninkovic S, Miljus-Djukic J, Neskovic M (1995) Genetic transformation of alfalfa embryos
and their clonal propagation through repetitive somatic embryogenesis. Plant Cell Tiss Org
Cult 42:255–260
37. Samac DA (1995) Strain specificity in transformation of alfalfa by Agrobacterium tumefa-
ciens. Plant Cell Tiss Org Cult 43:272–177
38. Schroeder HE, Schotz AH, Wardley-Richardson T, Spencer D, Higgins TJV (1993)
Transformation and regeneration of two cultivars of pea (Pisum sativum L.). Plant Physiol
101:751–757
39. Grant JE, Cooper PA, McAra AE, Frew TJ (1995) Transformation of pea (Pisum sativum L.)
using immature cotyledons. Plant Cell Rep 15:245–258
40. Pickardt T, Meixner M, Schade V, Scheider O (1991) Transformation of Vicia narbonensis via
Agrobacterium-mediated gene transfer. Plant Cell Rep 9:535–538
41. Karthikeyan AS, Sarma KS, Veluthambi K (1996) Agrobacterium tumefaciens mediated trans-
formation of Vigna mungo (L.) Hepper. Plant Cell Rep 15:328–331
42. Penza R, Lurquin PF, Filippone E (1991) Gene transfer by co-cultivation of mature embryos
with Agrobacterium tumefaciens :application to cowpea (Vigna unguiculata Walp.). J Plant
Physiol 138:39–43
Evaluation of Medicinal Plant
with Reference to Its Substitute
1 Introduction
Ayurveda is believed to be the oldest treatise on medical system, which came into
existence in about 900 B.C. The word Ayurveda is derived from the Sanskrit words
Ayur “meaning life” and Veda “meaning science.” Thus, Ayurveda literally means
science of life. According to the Indian Hindu mythology, there are four Vedas writ-
ten by the Aryans – Rig Veda, Sam Veda, Yajur Veda, and Atharva Veda. Rig Veda is
the oldest among the four Vedas written later than 1500 B.C. The Ayurveda is said
to be an Upaveda (part) of Atharva Veda, whereas Charaka Samhita (1900 B.C.) is
the first recorded book fully devoted to the concept of practice of Ayurveda. It
describes 341 plants and plant products for use in medicine. The next Ayurvedic
literature that has special emphasis on surgery was Sushruta Samhita (600 B.C.).
Another important authority of Ayurveda was Vagbhata of Sindh, presently in
Pakistan, who practiced Ayurveda in around seventeenth century A.D. His work
Ashtanga Hridaya is considered unrivaled for the principles and practices of medi-
cine. Madhab Nicane (800–900 A.D.) was the next important milestone and is the
most famous work on diagnosis of diseases as per the Ayurvedic concept. Bhava
Mishra of Magadha is the first writer on Hindu medicine whose treatise, Bhava
Prakasha, written around 1550 is held in high esteem by modern Ayurvedic practi-
tioners for its description of approximately 470 medicinal plants.
Side by side with the systematic development of medicines in ancient India,
there was also organized medical help in the form of hospitals and dispensaries and
a certain measure of health propaganda. During the Buddhist period, monks trav-
eled all over the country not only preaching religion and philosophy and dispelling
ignorance but also alleviating human suffering, King Asoka, and his edicts are
famous. But even before Asoka’s time, evidence of organized medical care was seen
in Charaka Samhita. Vivid descriptions are found about the location, building, and
personnel and amenities of hospital, Aturalaya; a maternity home, Sutikargriha; a
nursery, sisugriha; and a pharmacy. It should be remembered that Ayurveda is not
the science of health. Healthful living, prevention of disease, and personal and
social hygiene all come under its ambit and not merely the cure of diseases.
Indian medicinal plants are the essence of Ayurveda and Ayurvedic treatments.
When used judicially and clocking with the basic principles, they produce miracu-
lous effects. Their role cannot be confined to merely treating diseases but they are
also used being of the human body. Hence, Ayurvedic drugs are rightly called the
elixirs of life. Ayurvedic herbs played an important role in Ayurvedic treatment,
from the ancient time to the modern time.
Ayurvedic medicinal plants are classified according to their properties such as
Rasa, Guna, Virya, and Vipaka. (This is one of the simplest and most important clas-
sification I narrate, but there have so many classifications.) This classification helps
us how to manage/work medicinal plants to cure diseases. Ayurveda states that viti-
ation/abnormal increase in vata, pitta and kapha are main cause of disease. Vitiation
is due to the increase or decrease of similar properties. All the Dravya/substance has
properties (Guna).Vata, pitta and kapha have considerable increases in related
Doshas, so proper identification of Ayurvedic medicinal plants is important in the
field of treatment. Medicinal plants used in South India and North India are differ-
ent, even though their Sanskrit names are similar. However, they show the same
actions. We classified Ayurvedic medicinal plants according to their synonyms
which give exact idea or colorful pictures about medicinal plants. In Ayurveda, the
morphology of medicinal plants is planned in the form of synonyms.
Antioxidants have imperative anticipatory roles, not merely on detrimental
changes in the aroma and dietetic eminence of food, although on tissue damage in an
assortment of human diseases. Free radicals are generated all the way through cus-
tomary metabolism of drugs, ecological chemicals, and xenobiotic as well as endog-
enous chemicals, particularly stress hormones (adrenalin and noradrenalin).
Accumulated substantiation suggests that ROS can be scavenged during chemopre-
vention utilizing innate antioxidant compounds present in foods and medicinal plants.
High-performance thin-layer chromatography (HPTLC) is a semiautomatic
instrumental TLC. HPTLC is different from conventional TLC due to smaller par-
ticles (<10 μm) of adsorbent and less thickness of the applied layer (<150 μm). But
the most striking difference is that it has an automatic sample applicator which is
able to apply sample in micro liter amounts, an automatic development chamber
provided with solvent reservoirs, and a densitometer/scanner which scans separated
bands by measurement of optical density. Hence, the process is more efficient due
to small particle size and no manual errors.
The automatic sample applicator of HPTLC applies sample in the form of bands;
hence, it gives better separation and resolution especially for components having Rf
values like isomers of curcumin. Simple spot application in TLC causes mixing of
close Rf value components and chromatogram has a tailing appearance. HPTLC
enables simultaneous analysis of many samples in less time with better analytical
precision and accuracy.
Evaluation of Medicinal Plant with Reference to Its Substitute 929
2 Plant Description
Valeriana wallichii
2.1
2.2 Description
Indian valerian is an erect, perennial plant that grows to a height of 4 feet with pin-
nate, divided, and heart-shaped leaves. Pink or white flowers are found in clusters
on the leaf top. The roots are a hairy and spindly mass that is collected in the autumn
from two-year-old plants. The rhizomes are greenish-brown in color and hard and
tough internally. The herb is cultivated in Belgium, England, Eastern Europe,
France, Germany, the Netherlands, the Russian Federation, and the USA. The
medicinal plant is inhabitant to the Himalayas in Nagar, Minapin Glacier, and
Bultora Glacier in India. The herb is known as gilgiti valerian in Hindi and mushk
bala and risha wala in Urdu.
Valeriana officinalis
2.3
2.4 Description
3 Review of Literature
Nature has been a source of medicinal agents since time immemorial. The impor-
tance of herbs in the management of human ailments cannot be overemphasized. It
is clear that the plant kingdom harbors an inexhaustible source of active ingredients
invaluable in the management of many intractable diseases. Furthermore, the active
components of herbal remedies have the advantage of being combined with many
other substances that appear to be inactive. However, these complementary compo-
nents give the plant as a whole safety and efficiency much superior to that of its
isolated and pure active components.
Plants are potent biochemists and have been components of phytomedicines
since time immemorial; man is able to obtain from them a variety of industrial
chemicals. Plant-based natural constituents can be derived from any part of the plant
like bark, roots, leaves, flowers, fruits, seeds, etc., i.e., any part of the plant may
contain active components.
For the success of primary healthcare, the activity and the use of suitable drugs
is one of the prerequisite plants have always been a source source of medicaments,
either in the form of traditional preparations or as pure active principles. These
plants which are used as medicine to treat and cure diseases are called medicinal
plants. According to World Health Organization (WHO), a medicinal plant in one or
more of its organ, contains substances that can be used for therapeutic purposes or
precursors for the synthesis of useful drugs.
People in India and China are known to have used plants in organized healthcare
regime for over 5000 years. European herbal medicines blossomed in the Greco-
Roman era and remained in mainstream until six decades ago. The ancient civiliza-
tion of India, China, Greece, Arab, and other countries of the world developed their
own systems of medicine independent of each other but all of them were predomi-
nantly plant based. One of the oldest repositories of human knowledge, the Rig-
Veda (4500–4600 BC), mentioned the use of medicinal plants for the treatment of
diseases. In the long struggle to overcome the powerful forces of nature, human
beings have always turned to plants.
Nature has been a source of medicinal agents for thousands of years, and an
impressive number of modern drugs have been isolated from natural sources; many
of these isolations were based on the uses of the agents in traditional medicine. This
plant-based traditional medicine system continues to play an essential role in health-
care, with about 80% of the world’s habitants relying mainly on traditional medi-
cines for their primary healthcare.
Ayurvedic medicine is an ancient system of healthcare that is native to the Indian
subcontinent. It is presently used daily by millions of people in India, Nepal, and Sri
Lanka and indirectly through its being the major influence on Unani, Chinese, and
Tibetan medicine, in Tibet, and Pakistan too. The word Ayurveda is a tatpurusha
compound of the word ayus, which means life or life principles, and the word veda,
which refers to a system of knowledge. Thus, Ayurveda roughly translates as the
knowledge of life. According to Charaka Samhita, life itself is defined as the
Evaluation of Medicinal Plant with Reference to Its Substitute 933
combination of the body, sense organs, mind, and soul, the factor responsible for
preventing decay and death, which sustains the body over time and guides the pro-
cess of rebirth. According to this perspective, Ayurveda is concerned with measures
to protect ayus. This includes healthy living along with therapeutic measures that
relate to physical, mental, social, and spiritual harmony. Ayurveda is also one among
the few traditional systems of medicine to contain sophisticated system of surgery,
which is referred to as Salyachikitsa [2].
Herbs have been called part of nature’s pharmacy‖. Although their action can in
some ways be similar to modern drugs, herbal remedies are generally gentler and
safer. Many of the drugs used in conventional medicine are derived from herbs.
Herbalism uses the whole plant or whole parts of the plant, such as the leaves, the
flowers, or the roots. Using the whole plant helps in decreasing the side effects that
may occur when using isolated components. Herbs are plants that actually grown
fresh and purchased in dried form [3].
3.1 Valerianaceae
Valerian roots are acrid and bitter which are used as carminative and laxative and are
also used to treat blood diseases, burning sensation, cholera, skin diseases, throat
problems, and ulcers [4]. The use of extracts of the Valeriana roots and rhizomes for
sedation and to relieve sleep problems dates back to the eighteenth century [5], but
the exact composition of the preparations used was often not clear. According to
Suri et al. [6], its essential oil exhibited antimicrobial activity against pathogenic
bacteria and also exhibited potent antifungal activity against different human and
plant fungal pathogens.
In India, Valeriana has long been used in Ayurveda and Unani systems of medi-
cine, which describes its uses in skin diseases. Insanity, epilepsy, and snakebite are
considered to have remarkable sedative effects in nervous unrest, stress, and neural-
gia [4]. The plant is also used as cytotoxic. Roots of Valeriana wallichii are used as
an aphrodisiac and insecticide and to treat mental disorders [7].
In the search for the active substances of Valeriana, many compounds have been
isolated and identified during the last 120 years, but it is as yet uncertain which of
them are responsible for the recorded actions [8]. The most popular compounds, in
this connection, are the epoxy iridoids named valepotriates; their decomposition
products, the baldrinals; and the nonvolatile terpenoids grouped as valerenic acid
(VA) derivatives as well as some other members of the essential oil [8].
It is commonly known as Tagar which is an important ingredient of Ayurvedic
recipes for the treatment of nervous unrest and emotional problems. Besides, roots
provide commercially important essential oil used in perfumery. Morphologically,
it is a single species and is common in the Himalayan region (1220–2134 m) with
no subspecies or varieties. The investigations, however, revealed the existence of
chemically different forms (chemical races) within Valeriana wallichii DC on the
basis of chemical analysis of root extracts (essential oils and valepotriates)
934 T. V. Binu and C. B. Athira
responsible for the activity. Chemo type I is represented by maaliol (64.3%), while
type II possesses patchouli alcohol (40.2%) in their essential oils. Interestingly,
Charaka and Sushruta Samhita have also documented the existence of two types of
Tagar known as Pindtagar and Nata having medicinal properties [9]. It is a member
of the Valerianaceae family, consisting of about 200 species of Valeriana occurring
throughout the world [10] (Plate 1).
Valerian has been used as a medicinal herb since at least the time of ancient
Greece and Rome. Hippocrates described its properties, and Galen later prescribed
it as a remedy for insomnia. In medieval Sweden, it was sometimes placed in the
wedding clothes of the groom to ward off the “envy” of the elves. Sometimes people
put it in a tea. The Greek physician Dioscorides apparently recommended valerian
root to treat myriad disorders including heart palpitations, digestive problems, epi-
lepsy, and urinary tract infections. Valerian was recommended by Galen during the
second century as a treatment for insomnia. Valerian plants are as attractive as cat-
nip to cats, and it is rumored that the Pied Pipers secret to clearing the streets of
Hamlin was a store of valerian under his cloak. By the eighteenth century, valerian
was widely used as a sedative and to treat nervous disorders associated with a rest-
less digestive tract as well as the vapors in women. During World War I, valerian
was used to prevent and treat shell shock in frontline troops, and it was used during
World War II to help calm civilians subjected to air raids. Valerian was listed as a
sleep aid and anxiolytic in the US National Formulary until the 1940s. It fell into
disuse as more potent sedative-hypnotic pharmacologic agents became available.
Related species have been used in traditional Chinese medicine (TCM), Ayurvedic
medicine, and African herbal healing practices. Valeriana fauriei is used in tradi-
tional Chinese medicine and Japanese medicine as a sedative, a spasmolytic, and an
antidepressant. Valeriana capensis is used in African traditional medicine as a treat-
ment for epilepsy, hysteria, and nervous disorders. In the 1980s, valerian again
assumed a place of importance as a widely used nonprescription hypnotic and day-
time sedative, particularly in France, Belgium, Switzerland, Britain, Russia, and
Germany. Over 50 tons of valerians are sold each year in France alone. Adolescents
and young adults appear to be particularly attracted to valerian and other herbs that
affect the central nervous system. The German Commission E has given valerian
root a positive evaluation for use in states of restlessness. The European Scientific
Cooperative on Phytotherapy cites its indications as tenseness, restlessness, and irri-
tability with difficulty in falling asleep. Valeriana is used as medicine for insomnia,
anxiety, depression, menopausal symptoms, and stress and also used as sedative.
The unproven uses include absence of menstrual period, aches, acne, anorexia,
arthritis, etc. [11].
The genus Valeriana (family – Valerianaceae) comprises over 250 species, dis-
tributed throughout the world, and about 12 species are found in India [12]. The
genus is known for its popular name “valerian,” which is derived from the word
“valeo,” meaning to be strong, in the ninth century by Indian pharmacist and physi-
cian. In Latin, valerian is derived from the word “valere” which means to be well,
referring either to its aroma or its clinical effects. The genus is categorized as a
rasayana drug in traditional “Indian System of Medicine Ayurveda” and approved
as GRAS (generally recognized as safe) food ingredient in the USA [13].
Valeriana wallichii
3.2
Roots are acrid and bitter which are used as carminative and laxative and are also
used to treat blood diseases, burning sensation, cholera, skin diseases, throat prob-
lems, and ulcers [4]. Its essential oil exhibited antimicrobial activity against patho-
genic bacteria and also exhibited potent antifungal activity against different human
and plant fungal pathogens [6].
In India, Valeriana has long been used in Ayurveda and Unani systems of medi-
cine, which describes its uses in skin diseases. Insanity, epilepsy, and snakebite are
considered to have remarkable sedative effects in nervous unrest, stress, and neural-
gia [4]. The plant is also used as cytotoxic. Roots of Valeriana wallichii are used as
an aphrodisiac and insecticide and to treat mental disorders [7]. Valeriana wallichii
(VW) or Indian valerian is one of the several valerian species indigenous to the
temperate Himalayas found in greater parts of India [14].
936 T. V. Binu and C. B. Athira
Valeriana officinalis
3.3
Its roots and rhizomes have been used in traditional Chinese medicine for their
sedative and antispasmodic properties [15]. Aroma: When dried properly, Valeriana
officinalis L., s.l. has only a very faint characteristic, valeric acid-like aroma which
becomes stronger as it ages. Improperly dried or old material possesses a strong and
characteristic odor due to the enzymatic hydrolysis of esters of valepotriates (isova-
leric acid and hydroxyvaleric acid). Taste: It tastes mildly sweet and camphoraceous
with a slightly bitter and spicy aftertaste [16, 17] (Plate 2).
When dried, the whole rhizome is up to 50 mm long and up to 30 mm in diam-
eter, obconical to cylindrical, with an elongated or compressed base. It has a yellow-
ish- brown to dark brown exterior with a circular stem and leaf scars. The rhizome
contains numerous thick, light to dark brown rootlets which are located around a
thin ligneous cord. The root is longitudinally wrinkled and approximately 100 mm
long and 1–3 mm in diameter, almost cylindrical, and almost the same color as the
rhizome. In longitudinal section, the pith exhibits a central cavity transverse by
septa. The solons are 20–50 mm long and pale yellowish gray in color with promi-
nent nodes separated by longitudinally striated internodes. It is commonly sliced in
half for ease of cleaning. The rootlets, which contain the majority of the essential
Evaluation of Medicinal Plant with Reference to Its Substitute 937
oil, are brittle and break in short, horny fractures and are whitish or yellowish
internally.
The major groups of constituents in this plant are valepotriates and sesquiter-
penes. It is not yet clearly understood which components of Valeriana officinalis are
responsible for the therapeutic properties [18]. The studies of the Chemical Analysis
and Biological Activity of the Essential Oils of Two Valerianaceous Species from
China: Nardostachys chinensis and Valeriana officinalis concluded that the isolated
essential oils could be used for future development of antimicrobial and antioxidant
agents [19].
Valeriana officinalis has been known as one of the highly effective sedatives and
restorative remedies for the nervous system. Early uses of valerian for its bitter and
aromatic qualities are reported. Related plants, spikenard derived from Nardostachys
jatamansi DC, are mentioned in the Bible. The Greek physician and pharmacist
Galen (131–201 AD) was probably the first to allude to the sedative qualities of
valerian, while the English doctor John Hill in the mid-eighteenth century first used
it therapeutically as a sedative. Valerian was chemically analyzed in the nineteenth
century and by the end of the century, the pharmaceutical texts included the uses of
valerian in a way almost as accepted today. Valerian has been prescribed and used
as the perfect herbal tranquilizer in World War I to treat soldiers suffering from
shell shock.
938 T. V. Binu and C. B. Athira
4 Phytochemistry
extract. Plant chemicals are regarded as secondary metabolites because the plants
that manufacture them may have little need for them. They are synthesized in all
parts of the plant body: barks, leaves, stems, root, flowers, fruits, seeds, etc., i.e., any
part of the plant body may contain active components.
The chemical components identified in the solvent extract of Valeriana wallichii
constitute cyclopentaneacetaldehyde, quinoline, aristolene, cyclohexane,
2,4-diisopropenyl methvinyl, cyclopentane,1-(3-methylbutyl), tricyclo[5.1.0.0(2,4)
oct-5-ene-5-propanoic acid, ascorbic acid 2,6-dihexadecanoate, 9,12-octadecadienoic
acid, pentadecanoic acid, 4-hexadecyl ester, and valeric acid as the major
constituents.
Preliminary phytochemical analysis confirmed presence of carbohydrate, phe-
nols, flavonoids, tannins, alkaloids, steroids, glycosides, phytosterols, and saponins.
Quantitative analysis showed presence of about 7.6% phenols, 1.3% flavonoids,
1.1% tannins, and 3.5% saponins. Spectroscopic study indicated that the isolated
compound is a phenolic compound: 3,4,5-trihydroxybenzoic acid.
The phytochemical screening of Valeriana wallichii revealed the presence of
flavonoids, reducing sugar, terpenoids, tannins, and phenolics. The therapeutic
action of the plant is due to the presence of major chemical constituent flavonoids.
This chemicals work with nutrients and fibers to form an integrated part of defense
system against various diseases and stress conditions. The result of preliminary
phytochemical analyses identifies the presence of different bioactive groups such as
terpenoids and phytosterols (in hexane extract), phenols and flavonoids (in ethyl
acetate), and phenols, flavonoids, and steroids (in ethanol extract) [22].
The Valeriana wallichii extract obtained was dried and used for qualitative anal-
ysis of different constituents like alkaloids, tannins, flavonoids, cardiac glycosides,
proteins, carbohydrate, reducing sugar, anthraquinone, and steroids in ethanolic
extract [23].
All plants produce chemical compounds as part of their normal metabolic activi-
ties. These phytochemicals are divided into (1) primary metabolites such as sugars
and fats, which are found in all plants, and (2) secondary metabolites, which are
found in a smaller range in plants, serving a more specific function. For example,
some secondary metabolites are toxins used to deter predation and others are phero-
mones used to attract insects for pollination. It is these secondary metabolites and
pigments that can have therapeutic actions in humans and can be refined to produce
drugs, for example, inulin from the roots of dahlias, quinine from the cinchona,
morphine and codeine from the poppy, and digoxin from the foxglove. Toxic plants
even have use in pharmaceutical development. Plants synthesize a bewildering vari-
ety of phytochemicals, but most are derivatives of a few biochemical motifs.
940 T. V. Binu and C. B. Athira
Total phenols in the methanolic extract of roots and aerial parts were determined by
the Folin-Ciocalteu colorimetric method described by Singleton and Rossi [24].
Rhizomes and roots contain cyclopentapyrans, acacetin-7-0-rutinosides, linarin-
iso-valerinate, and 4-methoxy-8-pentyl-1-naphthoic acid [21]. Flavonoids in the
methanolic extract of plant were determined by aluminum chloride colorimet-
ric method.
Two new flavone glycosides, acacetin 7-0-sophoroside and acacetin 7-0-(6-0-l-
rhamnopyranosyl)-sophoroside, were isolated from the rhizomes and roots of
Valeriana wallichii [25]. Total phenolic content of extracts was determined by the
Folin-Ciocalteu method [26].
A UV-Vis spectrometer (U-2001, Hitachi, Japan) is used for the quantification of
total phenols, when comparing with root samples of Valeriana wallichii from wild
individuals, a significantly higher (P < 0.05) total phenols (13.05 mg/g dry weight
gallic acid equivalent) [13].
The total phenols were determined calorimetrically by using the Folin-Ciocalteu
method; 2.5 ml of tenfold diluted Folin-Ciocalteu reagent, 2 ml of 7.5%sodium
carbonate, and 0.5 ml of phenolic extracts were mixed. After heating at 45 °C for
15 minutes, the absorbance was measured at 765 nm against a blank. The phenolic
content was expressed as gallic acid equivalents/g dry weight of sample [26]
(Table 3).
6 Chromatographic Analysis
Valerenic acids and valepotriates are considered among the constituents responsible
for the sedative effect of roots and rhizomes of Valeriana species. Packed column
SFC/UV (p-SFC) was applied to the analysis of valerenic acids and valepotriates
extracted from the underground parts of Valeriana officinalis. The separation of
these compounds by p-SFC was optimized by varying the conditions (i.e., columns,
column coupling and stationary phase coupling, density programs, types of modi-
fiers, and modifier programs). A CN-stationary phase using supercritical carbon
dioxide modified with methanol/water (95:5) as mobile phase was successful in
separating both valerenic acids and valepotriates. The qualitative and quantitative
results of p-SFC are comparable to HPLC for both valerenic acids and valepotriates,
but analyses are faster with p-SFC than with HPLC [27].
India has rich wealth of important medicinal flora and its varied climate is ideally
suited for the cultivation of medicinal plants. The quality of the plant-derived medi-
cine is a matter of great concerns as the utilization of plant materials for cure of
infections and chronic human diseases is increasing. One of the major drawbacks in
the popularization of plant-based drugs is the inconsistent quality of the formulated
preparations. High-performance thin-layer chromatography, high-performance liq-
uid chromatography, chemical fingerprinting, and establishment of their library for
the commercially important medicinal and aromatic plant materials will help drug
industries and various institutions for the quality evaluation of their products to
complete in the world trade market of medicinal and aromatic plants. HPTLC and
HPLC are most commonly used modern tools for quality testing of raw materials
and finished products.
Modern thin-layer chromatography is a powerful, reliable, and cost-efficient
method for qualitative and quantitative analysis; HPTLC has a wide range of utility
in the field of plant-based products. In addition to qualitative detection, it provides
semiquantitative information on the main active constituents of plant preparations
[28]. Raaman [29] had made studies about the theoretical principles of chromatog-
raphy. High-performance thin-layer chromatography (HPTLC) is a sophisticated
and automated form of TLC, and HPTLC is the fastest of all chromatographic meth-
ods. HPTLC can analyze about 100 samples of five to ten different types per shift.
HPTLC is a visual technique, where the chromatogram is visible.
The FST and TST are widely used models of depression for screening new anti-
depressant drugs. Three extracts of Valeriana wallichii DC (Valerianaceae) rhizome
and fluoxetine were studied for antidepressant-like activity in two behavioral mod-
els, namely, the forced swim test (FST) and the tail suspension test (TST). It is sug-
gested that the antidepressant-like action of this herbal plant is not contingent upon
its terpenoid constituents.
TLC is a relatively cheap but powerful technique used to screen plant extracts for
the presence of different types of phenolic compounds. The relative position or Rf
values of the phenolic spots can be used as a first indication of phenolic compounds.
Plates can be sprayed with various reagents to detect the phenolic compounds. The
942 T. V. Binu and C. B. Athira
Fecl3- K3Fe (CN) 6 spray reveals all phenolics as blue spots. Rhodamine B spray is
used for polyphenols and flavanols by pink spot. Saturated KIO3 reveals gallic acid
and galloyl esters by brown or orange brown spots. One percent AlCl3 solution in
ethanol by yellow fluorescence reveals flavonoid and blue spot reveals phenolic
acids [30].
The pharmacological effects of Valeriana wallichii (VW) aqueous root extract
on sleep-wake profile and level of brain monoamines on Sprague-Dawley rats.
According to the study, it can be said that Valeriana wallichii water extract has a
sleep quality improving effect which may be dependent upon levels of monoamines
in the cortex and brainstem. It serves as an herbal therapeutic intervention to improve
sleep quality, i.e., increased deep sleep [31].
Gallo et al. [32] studied the pharmaceutical performance of the prepared
Valeriana officinalis dry extract through the design of tablets. The manufactured
tablets showed good compactability, friability, hardness, and disintegration time.
Those containing a disintegrate (Avicel PH 101) exhibited the best pharmaceutical
performance, having the lowest disintegration time of around 40 seconds.
Sung et al. [33] investigated the effect of valerian root extract and its major com-
ponent, valerenic acid, on memory function, cell proliferation, neuroblast differen-
tiation, serum corticosterone, and lipid peroxidation in adult and aged mice.
High-performance thin-layer chromatography fingerprint profiling was per-
formed to confirm the presence of various phytocompounds. The biomarker com-
pound was isolated by column chromatography method and characterized by
UV-visible, H 1-NMR, C 13-NMR, and mass spectroscopy methods.
Physicochemical analysis showed the presence of about 18% water-soluble com-
ponents and about 14% alcohol-soluble components .
Lovelyn and his co-workers studied the acute and chronic (90 days) oral toxicity
of Valeriana wallichii rhizome hydroethanolic extract in Swiss albino mice. From
this work, it could be concluded that Valeriana wallichii rhizome hydroethanolic
extract didn’t exhibit mortality, morbidity, or any other neurologic, hematologic, or
biochemical adverse effects apart from sedation which is the extension of their
known pharmacological activity, after single oral dose of 2000 mg/kg bw (14 days
of observation) or after once daily 200 mg/kg, 600 mg/kg, and 1800 mg/kg oral
treatments for 90 days in healthy adult Swiss albino mice [34].
The levels of valepotriates, dihydrovalepotriate, and acetyl-valepotriates from
Valeriana jatamansi Jones, Valeriana officinalis L., and Valeriana officinalis L. var.
latifolia Miq were obtained by using the reverse-phase high-performance liquid
chromatography (RP-HPLC) method. The highest levels were observed in Valeriana
Jatamansi Jones, followed by Valeriana officinalis L., and Valeriana officinalis
L. var. latifolia Miq. In addition, the content of iridoids varied significantly among
different parts and habitats.
The result of TLC of an alcoholic extract of Valeriana wallichii roots and its
extract was done in order to separate out compounds as possible. According to the
study, the Rf value of the given spot was found to be of 0.16, 0.48, and 0.84, respec-
tively, which come under the range of the Rf value of valerenic acid. The reported Rf
value for valerenic acid is 0.48.
Evaluation of Medicinal Plant with Reference to Its Substitute 943
UV and FTIR analysis of Valeriana wallichii root extract was carried out. The
result of FTIR analysis shows the identification of various functional groups present
in the extract. The absorption frequency of Valeriana wallichii root ethanolic extract
(C=S Stretching), C-O Stretching (Alcohols), C-O Stretching (Phenols), C=C
stretching (Aromatic), (N-H Bending), C=N stretching. The result of ultra-visible
spectrum detecting absorbance maxima of the phytochemical component present in
drug extract [23].
The GC-MS analysis of essential oils from cultivated Valeriana officinalis L. var.
latifolia Miq. identified six compounds, bornyl acetate (60.19%), (−)-acetic acid
Rhodomyrtus enol ester (3.87%), α-terpinyl acetate (1.55%), acetyl carene (1.68%),
α-selinene (26.07%), and (Z,E)-α-farnesene (1.56%), comprising 94.92% of the
total content. Cultivated Valeriana officinalis L. var. latifolia Miq. consisted of a
higher number of simple components, which was predominated by bornyl acetate
relative to that of wild Valeriana officinalis L.
Approximately 0.5–2.0% of Valeriana spp. consists of essential oils by gas
chromatography-mass spectrometry (GC-MS) analysis, which varies with species,
climate, and growing environment. Valerian plants from high-altitude fertile and
sandy soil have significantly higher essential oil content and yield similar to that of
biennials compared to annuals. Valerian plants that produce a higher amount of
essential oil are cultivated between September and November, although the content
of essential oils decreases with longer periods of propagation.
A total of 150 compounds have been identified in the essential oils of valerian
plants, mainly including monoterpenes and sesquiterpenes. Most monoterpenes,
namely, borneol, bornyl acetate, and isobornyl acetate, exhibit various bioactivities.
Around 30 sesquiterpene have also been detected in the valerian essential oils.
These have been classified to be of the guaiane type and valerian type. Despite the
low contents of these essential oils, their biological activities have drawn the atten-
tion of researchers around the world.
7 Antioxidant Activity
Recent interest in antioxidants due to their involvement in the health benefit had led
to the development of a number of antioxidant capacity assays. Plants contain high
concentration of numerous redox active secondary metabolites or antioxidants, such
as ascorbic acid, carotenoids, glutathione, tocopherols, tocotrienols, and enzyme
with high antioxidant activity to help them protect against hazardous oxidative dam-
age. The simple definition of an antioxidant as described by Halliwell is a molecule
which, when present in small concentrations compared to that of an oxidizable sub-
strate, significantly delays or prevents the oxidation of the substrate‖ [35].
Shariq proposed that the hepatoprotective activity of the Valeriana wallichii
extracts could be due to its antioxidant activity [36]. The antioxidant activity of
methanolic extract could be attributed to the presence of rich amount of polyphenols
944 T. V. Binu and C. B. Athira
and flavonoids. Essential oil of Valeriana jatamansi roots showed moderate antioxi-
dant activity.
The antioxidant activity of essential oil and supercritical fluid extracts was evalu-
ated in DPPH radical, superoxide radical, and hydroxyl scavenging assays. The
reducing power of those oil and extracts was also studied. Essential oil and super-
critical fluid extracts gave comparable DPPH, superoxide radical scavenging, and
reducing power activity, and no much deviation among various activities was
observed. However, when we consider overall antioxidant assays, 200 bar extract of
valerian is the best among essential oils and extracts.
Antioxidants have imperative anticipatory roles, not merely on detrimental
changes in the aroma and dietetic eminence of food, although on tissue damage in
an assortment of human diseases. Cellular smashup or oxidative injury arising from
free radicals or reactive oxygen species (ROS) nowadays appears as the elementary
mechanism underlying a number of human beings with neurodegenerative disor-
ders, viral infections, diabetes, inflammation, and autoimmune pathologies along
with digestive system disorders. Free radicals are generated all the way through
customary metabolism of drugs, ecological chemicals, and xenobiotic as well as
endogenous chemicals, particularly stress hormones (adrenalin and noradrenalin).
Accumulated substantiation suggests that ROS can be scavenged during chemopre-
vention utilizing innate antioxidant compounds present in foods and medici-
nal plants.
The methanolic extract of Valeriana wallichii was used in the present study and
also screened for the presence of phytochemicals, viz., alkaloids, flavonoids, tan-
nins, saponins, glycosides, etc. and their effect on 2,2-diphenyl-1-picryl-hydraxyl
radical (DPPH) which was used to determine the free radical scavenging activity.
8 Anxiolytic Effects
The use of extracts of the Valeriana roots and rhizomes to cause sedation and relieve
sleep problems dates back to the eighteenth century [5]. The anxiolytic effect of
valerian root extract, valerenic acid, and concentrations of valerenic acid and GABA
isolated in extract using the elevated plus maze. This method has been often used by
researchers to test the anxiolytic properties of drugs and is an effective measure of
animal anxiety [37].
A promising approach to detect new active substances in Valeriana extracts con-
sists in searching for the presence of ligands for the principal brain receptors pre-
dominantly associated with anxiolytic, sedative, and/or sleep-enhancing properties
[38]. There is growing reason to believe that valerian root may be an effective alter-
native to the traditional anxiolytics, which often produce such aversive side effects
such as nausea, tremor, and addiction [39]. Marder et al. [38] reported the identifica-
tion in Valeriana officinalis of the flavone glycoside linarin (LN) and the discovery
that it has, like HN, sedative and sleep- enhancing properties that are potentiated by
Evaluation of Medicinal Plant with Reference to Its Substitute 945
9.1 Plant Materials
9.2 Methods
The present study was carried out in the Research and Development (R & D)
Department of Oushadhi. The description of the method used for the study is
detailed below.
The plant materials used for this study consisting of dried rhizome of Valeriana wal-
lichii and Valeriana officinalis belonging to the family Valerianaceae were col-
lected from the Oushadhi premises. The rhizome was identified and authenticated
by a botanist of the Research and Development (R & D) Department of Oushadhi.
The dried samples of Valeriana wallichii and Valeriana officinalis were stored in
airtight containers. A portion of the dried samples were pulverized into powder by
using a mixer grinder.
946 T. V. Binu and C. B. Athira
9.2.3 Preparation of Extracts
The rhizomes of Valeriana wallichii and roots of Valeriana officinalis are examined
macroscopically. To compare the external characteristic features of the samples [42]:
10.1 Microscopical Analysis
10.1.1 Powder Microscopy
Procedure: Place the powered material on the slides. Add one to three drops of
glycerin-safranin mixture; if necessary stir with a fine-pointed needle to distribute
the testing agent evenly. Add a coverslip. Remove any excess liquid that may
exclude from under the coverslip by blotting around its edges gently with a filter
paper. Examine the slide under the microscope.
Hand selection of rhizome and roots of Valeriana wallichii and Valeriana officinalis
were cut using a sharp blade. Thin transverse selections were stained in a safranin
and then fast green, passed through alcohol grades for dehydration, and then
mounted in a DPX. Observations were taken from these sections using light micro-
scope. Special identifying features of the plant parts were studied and identified.
And prepare the permanent sides of plant parts.
Procedure for permanent slide preparation:
I. Collect the plant material and wash the material.
II. Section the material using a new blade.
III. Stain the specimen using alcoholic safranin for 20 minutes.
IV. Pass the specimen through the alcoholic series for 5 minutes (50%, 70%, 75%,
80%, 90%, and 95%).
V. Dip the specimen in the following chemicals:
(a) Isopropyl (10 seconds)
(b) Fast green (1 seconds)
(c) Clave oil (3 minutes)
(d) Xylene (5 minutes)
Evaluation of Medicinal Plant with Reference to Its Substitute 947
VI. Mount the section using DPX (using a new slide and coverslip).
VII. Examine the slide under the microscope (Peter Gray, 1958).
All the extracts of Valeriana wallichii and Valeriana officinalis were subjected to
qualitative tests for the identification of various active constituents. The following
are the reagents used for the phytochemical screening:
1. Test for alkaloids
(a) A small portion of the extracts, viz., chloroform, ethyl acetate, and ethanol,
was stirred with few drops of dil. Hydrochloric acid and filtered. To the fil-
trate, Dragendorff’s reagent
(potassium bismuth iodide solution) was added and an orange brown pre-
cipitate indicates the presence of alkaloids.
(b) To the filtrate, Mayer’s reagent was added and a cream precipitate indicated
the presence of alkaloids.
(c) Measured 2 ml of various extract solutions was then mixed with dilute
hydrochloric acid and 0.1 ml of Wagner’s reagent. Formation of reddish-
brown precipitate indicated the positive response for alkaloid.
2. Tests for flavonoids
To a portion of the various extracts, concentrated sulfuric acid was added. A
yellow coloration indicated the presence of flavonoids. The yellow coloration
disappeared on standing. Few drops of 1% aluminum chloride solution were
added to a portion of each fraction, and a yellow coloration indicated the pres-
ence of flavonoids.
3. Test for reducing sugars
Five ml of the extract is mixed with 5 ml of Fehling’s solution and boiled for
5 minutes. Formation of brick red-colored precipitate demonstrated the positive
test for reducing sugar.
4. Test for glycosides
To 1 ml of all the plant extracts, equal volume of Fehling’s solution A and B
was added. The mixture is heated in a water bath. Brick red coloration indicated
the presence of glycosides.
5. Test for steroids (Salkowski test)
Concentrated sulfuric acid was added to 10 mg of different extracts dissolved
in 1 ml of chloroform. A reddish-blue color exhibited by chloroform layer and
green fluorescence by the acid layer suggested presence of steroids.
6. Test for saponins
About 10 ml of the extract was mixed with 5 ml of distilled water and shaken
vigorously for a stable-persistent froth. The frothing was mixed with three drops
948 T. V. Binu and C. B. Athira
of olive oil and shaken vigorously and then observed for the formation of an
emulsion.
7. Test for terpenoids
About 5 ml of the extract was treated with 2 ml of chloroform and the concen-
trated sulfuric acid was carefully added to form a layer. A reddish-brown color-
ation of the interface indicates the presence of terpenoids.
8. Test for amino acid
Different extract solutions were treated with ninhydrin at the range of 4–8.
Development of a purple color indicated the positive response for amino acids.
9. Test for tannins and phenolic
(a) Around 5 ml of extract solutions was allowed to react with 1 ml of 5% ferric
chloride solution. Greenish-black coloration indicated the presence of
tannins.
(b) Approximately 5 ml of the extracts was treated with 1 ml of 10% aqueous
potassium dichromate solution. Formation of yellowish-brown precipitate
suggested the presence of tannins.
(c) Accurately 5 ml of the extracts was treated with 1 ml of 10% lead acetate
solution in water. Yellow color precipitation indicated presence of tannins.
10.3 Physicochemical Analysis
1. Stationary phase : TLC preloaded with silica gel 60 F254 of 0.2 mm thickness.
2. Solvent system : Toluene/ethyl acetate/formic acid (5:4:1).
3. Volume of the test : 7 μl.
solution applied
4. Spotting TLC : Spotting on the TLC plate was done using a 1–2 cm capillary tube from
plates the bottom of the plate.
5. Developing the : After spotting, the plates were kept inside the chamber in ascendant
plate position. After developing, the plate was taken from the chamber and let
the solvent be vaporized.
6. Detection spot : For the detection of spot, a TLC apparatus is used. The plate kept in the
apparatus containing flow of UV rays and color was analyzed.
7. Calculation of RF : RF value was calculated by using formula: Rf=DSU/DSV; Where Rf is
value the retention factor DSU is the distance traveled by the solute DSV is the
distance traveled by the solvent.
12 High-Performance Thin-Layer
Chromatography (HPTLC)
12.1 Terpenoids
Total phenols in the methanolic extract of root and aerial parts were determined by
the Folin-Ciocalteu colorimetric method described by Singleton and Rossi [24].
Methanolic extract (0.25 ml) diluted with distilled water (2.25 ml) was mixed with
the Folin-Ciocalteu reagent (0.25 ml) and allowed to stand for reaction for 5 min-
utes. This mixture was neutralized by 7% sodium carbonate (2.5 ml) and kept in
dark at room temperature for 90 minutes. The absorbance of resultant blue color
was measured at 765 nm using UV-Vis spectrophotometer. A UV-Vis spectropho-
tometer (U-2001, Hitachi, Japan) is used for the quantification of total phenols.
Samples were diluted with 80% (v/v) methanol to obtain 20–80% reduction in
absorbance at 734 nm with respect to blank. A standard curve of various
Evaluation of Medicinal Plant with Reference to Its Substitute 951
concentrations of ascorbic acid is prepared in 80% v/v methanol for the equivalent
quantification of antioxidant potential with respect to ascorbic acid. Results were
expressed in mM ascorbic acid equivalent (AAE)/100 g dry weight (DW).
15 Results
15.1 Macroscopical Analysis
15.1.1
Valeriana wallichii DC
The roots are yellowish brown in color and are found in broken pieces, 5–6 cm
length, and 1–2 mm in width; fine lateral rootlets are also present. The fracture is
horny and the broken surface is yellow. The odor is aromatic and some plants are
pungent. The taste is camphoraceous and slightly bitter.
The rhizomes are yellowish to dull brownish in color; they are subcylindrical in
shape and consist of rhizomatous portion connected by short thick stolon formparts
(showed in plate). The rhizomatous portions are about 1–5 cm long and about
5–15 mm thick. They appear knotted due to the presence of many raised root scars,
952 T. V. Binu and C. B. Athira
chiefly on the sides and upon the lower surface, which together with the numerous
transverse grooves and ridges formed by the leaf scars gives a very rough surface to
those parts of the commercial drug. The fracture is horny and the broken surface is
yellow. The odor and taste resemble that of the root [Pharmacognosy of Indian Root
and Rhizome Drugs].
15.1.2
Valeriana officinalis L
15.2 Microscopical Analysis
15.2.1 Powder Microscopy
Valeriana wallichii DC
The powder from roots and rhizome is characterized by brown color. There is pres-
ence of starch, root hairs are slightly lignified, and cells of the cortical parenchyma
are axillary elongated. The length is about six to ten times the width (plate).
Valeriana officinalis L.
brown granular contents; fragments of the endodermis show sinuous walls. Calcium
oxalate is absent (Elizabeth Williamson [45]).
15.3 Anatomical Analysis
15.3.1 Valeriana wallichii DC
Roots The young roots are characterized by the absence of cork, while it is present
in mature roots. The root hairs present in young roots and very much in length from
20–24 and 12–10 micro in width. The exodermal layers turn red with the use of
Sudan III and the cells are suberized and lignified. The cortex consists of 32 layer
cells; the outer three to four layers just within the exoderm are mostly collenchyma-
tous without any intercellular space and somewhat smaller in size than the cells in
the middle part of the cortex. Excepting those immediately outside the endodermis,
which are about the same size as the collenchymatous cells but are parenchymatous
in nature and with small intercellular space. Starch is present in the cortex. The
endodermis consists of one layer of prominent cells. Some of the cell’s dimension
is 16–28 μ × 136–225 μ. The pericycle consists of one to three layers of parenchy-
matous cells, with a dimension of 20–30 μ × 90 μ. Some of the cells contain tannoid
substances. The xylem is tetrarch to polyarch, having up to radially arranged bun-
dles. Vessels have a dimension of 205 μ–255 μ × 20–36 μ. The primary root contains
a little pith at the center. The stellar structure varies with age of the plant.
15.3.2 Rhizome
It is characterized by the presence of cork and other secondary tissues and the pres-
ence of a large pith. The stoloniform parts are different in structure from the rhi-
zomatous parts, chiefly in having no roots. Secondary development is well marked
and the number of vascular bundles is greater than in the rhizomatous parts.
15.3.3
Valeriana officinalis L.
The young roots are characterized by the absence of cork, while it is present in
mature roots. The root hairs are also present. The cortex consists of three to four
outer layers just.
within the exoderm which are mostly collenchymatous without any intercellular
space and somewhat smaller in size than the cells in the middle part of the cortex.
Starch is present in the cortex. Endodermis consists of one layer of prominent cells.
Some of the cells contain tannoid substance.
Evaluation of Medicinal Plant with Reference to Its Substitute 955
16 Phytochemical Evaluation
The HPTLC analysis of the terpenoid content of Valeriana wallichii and Valeriana
officinalis is carried out by using lupeol as the standard. The RF value and color of
the band obtained at wavelength of 366 nm are given in Table 4.
After derivatization using the Liebermann-Burchard reagent, major bands may
be present such as triterpenes, steroids, saponins, and bitter principles. The
HPTLC sample profiling of lane 1 showed five spots. The RF values of lane 1
(Valeriana officinalis) which are 0.13, 0.41, 0.44, 0.62, and 0.66 are shown in
Plate 3. The RF values of lane 2 (Valeriana wallichii) which are 0.12, 0.2, 0.44,
0.46, and 0.44 are shown in Plate 3. The values showed variations. Lane 2 has
large variation with RF value of 0.2 of brown-colored band when compared to lane
1 and lane 3 (STD).
The two samples of Tagara were treated for their antioxidant activities. Among
them, Valeriana wallichii has the higher antioxidant activity with 97.85% for
414.8 ppm when compared to the antioxidant activity of Valeriana officinalis with
97.42% for 414.8 ppm. Table 5 showed the details about the antioxidant activities of
Tagara with its different species.
16.2.1 Properties (Fig. 3)
Plate 3 Powder
microscopy. (a) Valeriana
officinalis L., (b) Valeriana
wallichii DC
17 Discussion
17.1.1 Macroscopic Analysis
17.1.2 Microscopic Analysis
The powdered forms of Valeriana wallichii and Valeriana officinalis are visually in
two different colors. A brownish color shows the Valeriana wallichii and a cream
white color shows the Valeriana officinalis. In Valeriana wallichii, starch is present,
root hairs are slightly lignified, and cells of the cortical parenchyma are elongated
axillary (Plate 5), whereas in Valeriana officinalis, vessels have reticulate thicken-
ing or bordered pits and fibers are occasional, lignified with simple pits (shown in
Plate 6), and lignified cells of tegumentary tissue from rhizome with brown granular
958 T. V. Binu and C. B. Athira
Plate 4 TLC profiling. (a) Valeriana wallichii DC, (b) Valeriana officinalis L.
Evaluation of Medicinal Plant with Reference to Its Substitute 959
Plate 5 Phytochemical
evaluation. (a) Valeriana
wallichii L., (b) Valeriana
officinalis L.
960 T. V. Binu and C. B. Athira
17.1.3 Phytochemical Analysis
Flavonoids are efficient quenchers of singlet oxygen and could be valuable anti-
oxidants in systems under oxidative stress, particularly if a flavonoid-rich diet was
previously consumed [51].
It is well known that rutin, an active flavonoid compound, possesses potent anti-
oxidant properties against oxidative stress. Rutin (50 μm) blocked apoptosis in
human umbilical vein endothelial cells through decreasing reactive oxygen species,
increasing glutathione, restoring DeltaPsim, and protecting DNA damage.
The existence of flavonoid glycosides with sedative and sleeping-enhancing
properties in Valeriana demonstrates the existence of potentiating effects in its
extracts. The suspected presence of synergic effects in Valeriana [52] has been sub-
stantiated by these findings and brought to the fore for future clarification of the
mechanisms involved [53].
Glycosides are molecules in which a sugar is bound to a noncarbohydrate moi-
ety, usually a small organic molecule. Glycosides play numerous important roles in
living organisms. Many plants store chemicals in the form of inactive glycosides.
These can be activated by enzyme hydrolysis, which causes the sugar part to be
broken off, making the chemical available for use. Many such plant glycosides are
used as medications. In animals and humans, poisons are often bound to sugar mol-
ecules as part of their elimination from the body. An example is the cyanoglycosides
in cherry pits that release toxins only when bitten by an herbivore. The flavonoids
are present in all extract of Valeriana species. Hence, both of them have antioxidant
property and flavonoid can protect the DNA damage. Steroids are in the methanolic
extract of Valeriana officinalis. Steroids have anti-inflammatory property. Steroids
are used as the main treatment for certain inflammation of blood, inflammation of
muscles, etc. They may also be used selectively to treat inflammatory conditions
such as rheumatoid arthritis, lupus, or gout.
Terpenoids were important secondary metabolites, because they are used as anti-
microbial, antifungal, anti-parasitic, antihyperglycemic, and anti-inflammatory
agents. They have immunomodulatory properties. Valeriana wallichii and Valeriana
officinalis have high medicinal values due to the presence of various secondary
metabolites.
17.1.4 Physicochemical Analysis
6.31, 33.2, and 21.3 are the total ash, acid-insoluble ash, water-soluble ash, alcohol-
soluble extractive, and water-soluble extractive values, respectively.
The phytochemical parameters of Valeriana wallichii and Valeriana officinalis
show similarity in their values (Fig. 4). From this study, it shows that the physico-
chemical properties of Valeriana wallichii and Valeriana officinalis are similar.
964 T. V. Binu and C. B. Athira
17.1.5 Chromatographical Analysis
Thin-Layer Chromatography
In Valeriana wallichii, blue color bands were present at Rf value of 0.51. Blue color
bands are present at Rf value of 0.27 in Valeriana officinalis. Small variations in
their band length are present. Hence, the TLC system developed can be effectively
used for differentiating two varieties of Tagara. A number of blue bands are present
in both samples, which shows similarity of chemical components of both samples
(Plate 7 and Table 8).
Plate 7 Macroscopic analysis. (a) Valeriana wallichii DC, (b) Valeriana officinalis L.
HPTLC
Terpenes are a large and diverse class of organic compounds, produced by a variety
of plants, particularly conifers, which are often strong smelling and thus may have
had a protective function. They are the major components of resin and of turpentine
produced from resin (the name “terpene” is derived from the word “turpentine”).
Terpenes are major biosynthetic building blocks within nearly every living creature.
Steroids, for example, are derivatives of the triterpene squalene. When terpenes are
modified chemically, such as by oxidation or rearrangement of the carbon skeleton,
the resulting compounds are generally referred to as terpenoids. Terpenes and terpe-
noids are the primary constituents of the essential oils of many types of plants and
flowers. Essential oils are used widely as natural flavor additives for food, as fra-
grances in perfumery, and in traditional and alternative medicines such as aroma-
therapy. Synthetic variations and derivatives of natural terpenes and terpenoids also
greatly expand the variety of aromas used in perfumery and flavors used in food
additives. Vitamin A is an example of a terpene. The fragrance of rose and lavender
is due to monoterpenes. The carotenoids produce the red, yellow, and orange colors
of pumpkin, corn, and tomato, respectively.
Valeriana officinalis has more terpenoid content than Valeriana wallichii.
Valeriana officinalis shows Rf values of 0.13, 0.14, 0.44, 0.62, and 0.66 with color
bands of gray, gray, green, gray, and green, respectively. Valeriana wallichii shows
Rf values of 0.12, 0.2, 0.44, 0.46, and 0.44 with color bands of green, brown, green,
blue, and blue, respectively. Here lupeol is used as the standard with a wavelength
of 0.75 with green band. Valeriana officinalis shows nearest value to lupeol. Hence,
it contain more terpenoids than Valeriana wallichii.
18 Antioxidant Activity
19 Summary
merely responsible for the activity of the plant. The present study on the evaluation
of Valeriana wallichii for the preliminary phytochemical could be used as the diag-
nostic tool for the standardization of medicinal plant. Thus, our study may be an
important landmark in the correct identification of Valeriana wallichii.
The antioxidant activity and terpenoid content are greater in Valeriana officinalis
than in Valeriana wallichii. The chromatographic analysis, phytochemical analysis,
and physicochemical analysis show similarity in the chemical constituents and
compound of both species of Valeriana. Hence, it can be used as good substitute in
place of Valeriana wallichii.
References
18. Letchamo W, Ward W, Heard B, Heard D (2004) Essential oil of Valeriana officinalis L. cul-
tivars and their antimicrobial activity as influenced by harvesting time under commercial
organic cultivation. J Agric Food Chem 52. ISSN: 3915-3919
19. Zhou JW, Zhao J, Liu H, Liu Z, Wang J, Han J, Yu Z, Yang F (2010) Chemical analysis and bio-
logical activity of the essential oils of two valerianaceous species from China: Nardostachys
chinensis and Valeriana officinalis. Molecules 15:6411–6422
20. Kemper KJ (1999) Valerian (Valeriana officinalis). Longwood Herbal Task Force. http://www.
mcp.edu/herbal/default.htm, 15 Dec 1999
21. Pandey A, Shukla YN (1993) Naphthoic acid derivative from Valeriana wallichii.
Phytochemistry 32:135
22. Rajalakshmy MR, Geetha G (2016) Isolation and identification of 5-O-caffeoyl quinic acid
from Valeriana wallichii. Asian J Chem 28(3):572–574
23. Dhongade HJ, Sahu V, Sawarkar HA, Sahu P, Sahu R, Patel D, Kashyap P (2016) Isolation
and characterization of Valerinic acid from Valeriana wallichii (Valerinaceae). IJBPAS 5(6)
24. Singleton VL, Rossi JA (1965) Colorimetry of total phenolics with phosphomolybolic acid-
phosphotunguntic and reagent. Am J Enol Viticult 16:144–158
25. Tang YP, Liu X, Biao Y (2003) Two new flavone glycosides from Valeriana jatamansi. J Asian
Nat Prod Res 5:257–261
26. Sayed A, Hossein G, Fereshteh M, Mehdi R (2012) Phenolic compounds and antioxidant
activity from saffron (Crocus sativus L.). Petal J Agr Sci 4:175–181
27. Bicchi, Binello A, Rubiolo P (2000) Packed column SFC/UV versus HPLC/UV analysis of
valerenic acids and valepotriates in extracts of Valeriana officinalis L. 11(3)
28. Ubadhyay A, Preeti S, Nayak M, Misra N, Afshankhan Dwivedi SK, Rao S (2006)
Standardisation techniques for medicinal and aromatic plants, 7–8
29. Raaman N (2006) Phytochemical techniques. New India Publishing Agency, New Delhi,
pp 19–77
30. Esmaeili N, Ebrahimzadeh H, Abdi K (2011) Determination of some phenolic compounds in
Crocus sativus L. corms and its antioxidant activities study. Pharmacogn Mag 7:74–80
31. Sahu S, Ray K, Yogendra Kumar MS, Gupta S, Kauser H, Kumar S, Mishra K, Panjwani U
(2012) Valeriana wallichii root extract improves sleep quality and modulates brain monoamine
level in rats. Phytomedicine 19:924–929
32. Gallo L, Ramírez-Rigo MV, Piña J, Palma S, Allemandi D, Bucalá V (2012) Valeriana offi-
cinalis dry plant extract for direct compression: preparation and characterization. Sci Pharm
80(4):1013–1026
33. Sung Min Nam A, Choi JH, Yoo DY, Kim W, Jung HY, Kim JW, Kang S-Y, Park J, Kim
D-W, Kim WJ, Yoon YS, Hwang IK (2013) Valeriana officinalis extract and its main compo-
nent, valerenic acid, ameliorate D-galactose-induced reductions in memory, cell proliferation,
and neuroblast differentiation by reducing corticosterone levels and lipid peroxidation. Exp
Gerontol 48:1369–1377
34. Joseph, Puthallath RE, Rao SN (2015) Acute and chronic toxicity study of Valeriana wallichii
rhizome hydro-ethanolic extract in Swiss albino mice. Asian J Med Sci 7(2)
35. Charles DJ (2013) Antioxidant properties of spices, herbs and other sources. Frontier Natural
Products Co-op, pp 509–516
36. Syed SN, Rizvi W, Kumar A, Khan AA, Moin S, Ahsan A (2014) A study to evaluate antioxi-
dant and hepatoprotective activity of aqueous extract of roots of Valeriana wallichii in CCl4
induced hepatotoxicity in rats. Int J Basic Clin Pharmacol 3(2):354–358
37. Pellow S, Chopin P, File S, Briley M (1985) Validation of open: closed arm entries in an
elevated plus-maze as a measure of anxiety in the rat. J Neurosci Methods 14(3):149–167
38. Marder M, Viola H, Wasowski C, Fernandez S, Medina JH, Paladini AC (2003) 6-
Methylapigenin and hesperidin: new valeriana flavonoids with activity on the CNS. Pharmacol
Biochem Behav 75:737–745
39. Stewart SH, Westra HA (2002) Benzodiazepine side-effects: from the bench to the clinic. Cur
Pharmaceut Des 8(1):1–3
Evaluation of Medicinal Plant with Reference to Its Substitute 969
40. Awad R, Levac D, Cybulska P, Merali Z, Trudeau VL, Arnason JT (2007) Effects of tradition-
ally used anxiolytic botanicals of enzymes of the g-aminobutyric acid (GABA) system. Can J
Phys Pharmacol 85(9):933–942. 5
41. Murphy K, Kubin ZJ, Shepherd JN, Ettinger RH (2010) Valeriana officinalis root extracts have
potent anxiolytic effects in laboratory rats. Phytomedicine 17:674–678
42. Neeraj T (2011) Quality standards of Indian medicinal plants. In: Medicinal plants unit. ICMR,
New Delhi, 9:96–98
43. Richa SS, Sharma ML (2014) Phytochemical investigation and anatomical study of three spe-
cies of Sida. Bio life 2(2):3–8
44. Graff A (1999) Valerian root. American Herbal Pharmacopoeia, p 2
45. Williamson E (1999) Valerian root (microscopy). American Herbal Pharmacopoeia, p 4
46. Wadood A, Ghufran M, Jamal SB, Naeem M, Khan A, Ghaffar R, Asnad (2013) Phytochemical
analysis of medicinal plants occurring in local area of Mardan. Biochem Analy Biochem
2(4):1000144
47. Yadav M, Chatterji S, Gupta SK, Watal G (2014) Preliminary phytochemical screening of six
medicinal plants used in traditional medicine. Int J Pharm Pharm Sci 6(5):539–542
48. Chic OI, Amom TT (2014) Phytochemical and antimicrobial evaluation of leaf-extracts of
Pterocarpus santalinoides. Euro J Med Plant 4(1):105–111
49. Radhika B, Murthy JVVSN, Grace DN (2013) Preliminary phytochemical analysis & antibac-
terial activity against clinical pathogens of medicinally important orchid Cymbidium aloifo-
lium (l.) sw. Int J Pharm Sci Res 4(10):3925–3931
50. Corradini E, Foglia P, Giansanti P, Gubbiotti R, Samperi R, Lagana A (2011) Flavonoids:
chemical properties and analytical methodologies of identification and quantitation in foods
and plants. Nat Prod Res 25:469–495
51. Morales J, Günther G, Zanocco AL, Lemp E (2012) Singlet oxygen reactions with flavonoids.
A theoretical – experimental study. PLoS One 7:e40548
52. Hobbs C (1989) Valerian: a literature review. Herbal Gram 21:19–34
53. Williamson EM (2001) Synergy and other interactions in phytomedicines. Phytomedicine
8:401–409
Biotechnology: Production of Natural
Bioactive Compounds from Leguminous
Plants and Disease Management
1 Introduction
Malnutrition is a severe and common issue in public health. The size and severity of
the issue and the functionality of the weaknesses in both physiological and socio-
economic terms need understanding and immediate intervention as a matter of
urgency [1, 2]. Having said that, the focus on their own narrow area of interest or
expertise, whether it is health care or food, discouraged development professionals
from using a truly holistic approach to dealing with the critical problem [3]. This
article is going to remedy this disparity and bring food-based practices back at the
forefront of conversation and facilitate its acceptance on a wider scale. In both
emerging and developed nations, micronutrient shortages occur and may be seen as
“secret hunger.” In developing nations, they occur in the sense of food poverty,
where total nutrition demands and food diversity prove to be the key problem.
Efforts to minimise micronutrient deficiency thus have to be made in the sense of a
projected 854 million population, 20 million under-five children subjected to
extreme malnutrition, and around one million kids die per year as a consequence of
malnutrition [4].
Iron deficiency is the most widespread micronutrient deficiency globally, which
affects the brain tissue. Iron deficiency anaemia is one of the six priority nutrition
measures for the Sustainable Development Targets. Malnutrition caused by a lack of
crucial mineral micronutrients such as iron (Fe) in their food is a major issue that
affects almost half of the world’s population. Also, in the absence of anaemia, iron
deficiency can cause fatigue and affect work efficiency [5]. Dietary iron absorption
is a dynamic mechanism that has received a lot of attention. Dietary iron is made up
of elemental iron as well as either haem or non-haem iron. The molecular processes
of absorption and bioavailability of these types vary significantly. Thus, both the
overall amount of iron and the form of iron in food are essential nutritionally [3].
Calcium blocks the synthesis of haem and non-haem iron during the initial iron
uptake into the enterocyte. Young women are particularly vulnerable to iron defi-
ciency as a result of menstrual and childbirth losses, according to the International
Journal of Science. The most important thing that raises a female’s risk of iron
deficiency is menstruation [6]. According to one report, even though volunteers ate
five portions of red meat and two portions of poultry per week, the recommended
dietary allowance (RDA) of iron for women was not easily met [7]. The reasons for
the variance among the Reference Dietary Intakes for iron and real iron consump-
tion have been proposed as a lack of availability, a lack of access to fresh foods, and
a lack of nutrition information and understanding.
The major reason for such high levels of malnutrition is insecurity and inade-
quate agricultural production. FAO is dedicated to alleviating the rising issue of
food poverty in the world, so that everybody will have access to healthy food [8].
This has been achieved by FAO by growing the provision of food-based services to
prevent starvation by enhanced intake of a sufficient and diverse diet. This illus-
trates the right to health, which means to ensure that everyone must have access to
a range of diets containing a variety of foods in order to have enough calories and
macronutrients [9].
1.1 Leguminous Seeds
Grain legumes are an excellent source of protein. Their exploitation is set to increase
as the world’s food needs increase. Furthermore, the positive benefits of legume
seed dietary consumption, which are the basis for several health statements, are cur-
rently being re-examined. Proteins and peptides are involved in the observed bio-
logical processes of legume plants, but their effects have not been fully revealed
[10–12]. Plant-based protein ingredients have gained prominence in the food indus-
try in recent years, as market habits have shifted away from the use of animal-
derived proteins (e.g., casein, whey, and egg) in favour of dietary choices based on
religious, moral, or health reasons, cost, and availability [13]. In general, legumes
and cereals are high in protein, carbohydrates, fibre, vitamins, minerals, and photo-
chemical. They are often eaten together in the diet as supplementary protein sources,
with legumes being high in lysine but low in methionine and cysteine, while cereals
are the opposite [14].
Legumes and cereals are mostly used in their natural flour form because they are
less costly than purified products like starches or protein isolates and concentrates,
or chemically processed flours. There is an increasing trend in incorporating legume
flours into standard cereal and legume-based foods such as pizza, chips, tortilla, and
noodles and extruded snack products. This is leading to growth in modern pulse
products. Going forward, legumes also show excellent opportunities in frozen
dough foods either as added flour or as fillings. Pulses in view of their nutrient
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 973
profile seem to be ideal for inclusion in designing snack foods, baby, and sports
foods [15, 16]. However, flours from different origins can behave differently and
have different effects on the nutrient composition of the product; thus, it is critical
to consider how these various flour ingredients react with one another [17]. Based
on their functionality and use, cereal and legume flours may be used. Soybean flour,
for example, is frequently added to foods to increase the protein content of the prod-
uct, while wheat flour comprises gluten, a protein that provides dough with the
special viscoelastic properties used for bread making. Water or oil-carrying capabil-
ity, solubility, foaming, and emulsion capacity/stability are all practical properties
of flour [18]. Because of the lack of uniform measurement protocols, variation in
raw material composition within the same form of crop seed due to genotype and/or
climate, and differences in seed processing technique, the functionality of legume
and cereal flours is difficult to compare in the literature (grinding, dehulling, and
defatting). Few researches have used both legumes and cereals in the same survey
in order to effectively and precisely to compare them [19].
In developed nations, legumes and cereals meet the majority of people’s energy
and protein needs. Even where the dietary iron content is sufficient, nutritional iron
deficiency is normal in these regions [20–22]. Iron bioavailability studies have
shown that most cereals, with the exception of highly processed wheat, absorb iron
poorly. Legumes contain more iron than cereals, but its bioavailability in humans
has not been thoroughly investigated. Recent studies show that soybeans and soy
protein materials have a poor level of iron absorption. However, since no clear com-
parisons of iron supply in soybeans and other legumes in humans have been
recorded, iron absorption from five widely consumed legumes was measured in
healthy volunteers [7].
Radio isotopic tests of iron absorption in human subjects were used to assess iron
bioavailability in a number of legumes. Soybeans, black beans, lentils, mung beans,
and split peas were made into soups and served to fasting subjects using the extrin-
sic tag system. The average percentage absorption ranged from 0.84% to 1.91%.
Absorption differences between means were not statistically important. According
to the findings, these five widely consumed legumes are all low in dietary iron
[23–24].
Legumes are well known for containing a variety of bioactive compounds, includ-
ing saponins, flavonoids, lectins, tannins, phytic acid, isoflavones, and others, which
contribute to their nutraceutical properties. The phenolics and flavonoid content of
highly pigmented and dark-coloured legume seeds is higher, which aids in antioxi-
dant function [25–27]. Legume seeds contain enzyme inhibitors such as alpha-
amylase, beta-glycosidase, and gamma-aminobutyric acid (GABA), making them
useful as a nutraceutical molecule. Green legume seeds are also high in nutraceuti-
cals. Legume seeds are usually eaten after being processed to increase nutritional
bioavailability by inactivating trypsin, growth promoters, and hemagglutinins [9].
974 S. Jha and B. Jain
2.1.1 Tannins
Because of their ability to bind proteins, tannins aid in the elimination of poisons
from the intestine. Tannins also aid in oral hygiene by inhibiting the growth of bac-
teria that cause tooth decay [1].
2.1.2 Flavonoids
2.1.3 Isoflavones
2.1.4 Phytic Acid
2.1.5 Saponins
2.1.6 Lectin
induce mitosis while inhibiting lymphocyte cap and patch development due to anti-
immunoglobulin. Con A exhibits similar behaviour to insulin in isolated fat
cells [19].
2.1.7 Protein
Pea, faba bean, and beans mature seeds produce 18–20% protein, while lupin and
soybean seeds contain 35–45% protein. The majority of the proteins present in
legume seeds are storage proteins with sedimentation coefficients of 7S and 11S
globulins. Some proteins in legume seeds have antifungal and antiviral function,
making them anti-HIV and antidiabetic. These proteins contain a number of impor-
tant amino acids that are good for human health [41]. The proteins found in Vigna
species have antifungal and antiviral properties. Ground bean lectin blocks polyga-
lacturonic acid’s hemagglutinating activity but not galacturonic acid or basic mono-
saccharides. It reduces the viability of hepatoma (HepG2), leukaemia (L1210), and
leukaemia (M1) cells while also inducing a mitogenic response in mouse spleno-
cytes. Since these proteins possess both of these features, they serve as an ideal
agent for the treatment of AIDS patients, with little adverse effects when compared
to pharmaceutical medications [42, 43].
2.1.8 Fibre
Legumes are a great source of fibre. Dietary fibre is derived from the part of plants
that is not digested by intestinal enzymes. Bacteria in the lower intestine can metab-
olise this and produce short-chain fatty acids. Fibre also lowers blood cholesterol
levels by binding with cholesterol in the human intestine. For diabetics, high-fibre
diets may increase serum lipoprotein levels, reduce blood pressure, and improve
blood glucose levels. Insoluble fibre increases the amount of waste content transient
time from the gastrointestinal tract [33].
2.1.9 Phytic Acid
Legumes’ phytic acid prevents colon cancer by iron chelation and inhibition of iron-
related carcinogenesis initiation and progression. Furthermore, because of its ability
to increase the activation of natural killer cells, which is consistent with suppressed
tumour occurrence, it may have potential therapeutic application in cancer [11, 44].
Soybean is a better source of vitamin B than cereals, but it lacks vitamin B12 and
C. Tocopherols, which are powerful natural antioxidants, are also found in soybean
oil. Soybean also contains 5% minerals. It has a high concentration of K, P, Ca, Mg,
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 977
and Fe. Soy ferritin has a high concentration of iron. Minerals, such as copper, zinc,
magnesium, and manganese, serve as cofactors in certain enzymatic reactions.
Vitamins E and C are known to act as antioxidants and to prevent vitamin A oxida-
tion in the gastric intestinal tract. Vitamin E also protects against cancer by inhibit-
ing the formation of carcinogens from precursor compounds. Vitamin K, on the
other hand, serves a functional purpose in the liver by acting as a blood clotting
agent. Folic acid, a B vitamin, greatly decreases the risk of neural tube defects
(NTDs) such as spina bifida in newborns [37, 45].
2.1.11 Photochemical in Legumes
2.1.12 Legume Fibres
Legumes are an excellent source of dietary fibre. Resistant starch, non-starch poly-
saccharides (cellulose, hemicelluloses, pectin, gums, and b-glucans), non-digestible
oligosaccharides, and lignin are examples of dietary fibres. Soluble fibre intake is
associated with lower serum total cholesterol (TC), LDL-C, and is inversely linked
to CHD mortality incidence. Dietary fibres can also help prevent obesity [46].
2.1.13 Antioxidant Activity
unique, such as some complex glycosides. Antinutrients in legume seeds are thought
to reduce protein and carbohydrate absorption. Since high temperatures during pro-
cessing inactivate the ANCs, the harmful effects of legume seeds are only found
after ingestion of raw and unprocessed seeds or flour. Most bean ANCs have an
impact on the digestive system, such as inhibiting digestive enzymes (e.g., protease
inhibitors), impairing hydrolytic functions and transport at the enterocyte site (lec-
tins), forming insoluble complexes (phytates, polyphenols), and increasing gas out-
put throughout the colon (beta-galactosides). The most well-known and widely used
protein inhibitors in legume seeds are trypsin inhibitors of both the Bowman-Birk
and Kunitz types, as well as amylase inhibitors present in chickpea. Most legumes,
such as kidney beans and grain beans, are also high in lectins. Legume seeds also
contain a variety of non-protein ANCs such as phenolic compounds, saponins, alka-
loids, phytates, and others that interfere with the biological use of their nutrients
[47]. Figure 1 shows benefits of phytochemicals.
Saponins have antimicrobial effects, which protect the body from fungi, bacteria,
and viruses. Simultaneously, they boost immune function by stimulating T-cell
development. They also act as antioxidants and scavengers of oxidative stress and it
is very useful in the management of coughs and the control of upper respiratory
soreness. Furthermore, plant-based saponins act as a natural heart tonic and have
been shown to prevent diabetes and fungi development [33].
Tannins have been shown to maintain functional and extreme haemostatic proper-
ties that aid in wound healing, improve irritated mucus membranes, and inhibit
bacterial growth, lowering blood pressure, lowering serum lipid levels, causing liver
necrosis, and modulating immune responses. This activity is accomplished by the
precipitation of proteins in bacteria, thus impeding the utility of proteins by bacteria
and, as a result, slowing protein production within the cells. The amount and type of
tannins used are important factors in these effects. Since tannins are also responsible
for a decrease in appetite, feed quality, and protein digestibility; foods high in tan-
nins are thought to be of poor nutritional value [48].
Flavonoids are important because they regulate and avoid tissue damage caused by
the presence of activated radical, or singlet oxygen organisms. Furthermore, flavo-
noids have a variety of biochemical properties. They function as vasodilators, reduce
swelling of body tissues, act as antioxidants and antimicrobial, and boost the
immune system’s efficacy [32].
Steroids are well-known essential cardiac stimulants. They have the ability to pro-
tect against mosquito and bacterial attacks. They are used in both cosmetics and
food, as herbal medicine to boost resilience. Because of their profound and efficient
inhibition of microbial growth, they are useful in conventional health services or
applications [40].
It is a soybean variety with a black seed coat that has long been used as a tonic food
and substance in oriental medicine. Because of its active peptide compounds, tradi-
tional Chinese medicine theory suggests that black soybean is beneficial in the treat-
ment of diabetes, asthma, antiageing, cosmetology, blood circulation, and other
diseases [7].
2.1.16 Pigeon Pea
Meat, starch, calcium, manganese, crude fibre, fat, trace elements, and minerals are
all abundant in this legume food. Pigeon pea seeds are made up of 85% cotyledons,
14% seed coat, around 1% embryo, and a mixture of dietary nutrients. The majority
of the seed proteins are found in the foetus, while the majority of the carbohydrates
980 S. Jha and B. Jain
are found in the cotyledons. It is both nutritious and therapeutic in nature. When
scorched seeds are mixed with coffee, they can help with headaches and vertigo,
while fresh seeds can help with male urinary incontinence [17]. Immature beans, on
the other hand, are used to cure kidney problems. Pigeon pea seed husks have anti-
oxidant and antihyperglycemic action, making them a promising sustainable option
for the production of hyperglycaemic nutraceuticals.
Chickpeas are in high demand all over the world due to their nutritious value.
Chickpea is an essential part in the diets of those who cannot afford animal proteins
in the semi-arid tropics. It is a great source in carbohydrates and protein, and the
quality of protein it contains is considered to be better than other pulses. Chickpeas
are low in cholesterol and high in calcium, dietary fibre (DF), vitamins, and miner-
als. Consumption of chickpeas has been shown to lower the incidence of chronic
diseases and improve health. Chickpea seed oil comprises a variety of sterols,
tocopherols, and tocotrienols. These phytosterols have been shown to have antibac-
terial, antiulcerative, antifungal, antitumor, and anti-inflammatory properties, as
well as a cholesterol-lowering impact [7].
Phaseolus is the world’s most essential food legume for human consumption. Its
seedlings are mostly composed of carbohydrates and are a rich source of nitrogen
and protein. It also has several elements found in the human body like calcium,
magnesium, potassium, arsenic, copper, iron, zinc, manganese, and sulphur.
This legume contains a high concentration of bioactive ingredients such as
enzyme inhibitors, lectins, phenolics, phytates, and oligosaccharides all of which
play biochemical functions in humans and animals. Among them are the observed
biological processes include antioxidant potential and cholesterol reduction. As a
result of the elimination of low-density lipoproteins, Phaseolus has a defensive
function against cardiovascular problems. It has also demonstrated anticancer activ-
ity due to the antimutagenic and antiproliferative properties of its phenolics, lectins,
and protease inhibitors. It also has antiobesity and antidiabetic properties due to the
presence of resistant starch and α-amylase inhibitor [3].
The nutritional value of legumes has been extensively researched. Legumes are high
in protein, fat, carbohydrates, nutrients, antioxidants, calcium, and water, as well as
being good source of thiamin (B1), beta-carotene (provitamin A), riboflavin (B2),
pyridoxine (B6), niacin, folic acid (folacin), pantothenic acid, ascorbic acid, and
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 981
vitamins E and K. According to recent research, there are many ways to increase the
supply of nutritious nutrients through good cooking method collection [9]. The
most popular methods for cooking legumes, according to these reports, are steam-
ing, roasting, boiling, baking, sautéing, microwaving, and pressure-cooking. Aside
from that, the authors considered factors related to traditional domestic production,
such as washing, peeling, cutting, slicing, and soaking in their studies. Such infor-
mation has been researched for specific vegetables (broccoli, potato, and onion) and
legumes (beans and peas). Many studies have revealed major variations in cooking
techniques and investigated the effect of cooking on in vitro bile acid binding by
various vegetables. Bile acid binding has been found to lower cholesterol levels in
the blood, thus decreasing the risk of heart disease. In their first research, they dis-
covered that steam cooking increased bile acid binding in beets, eggplant, aspara-
gus, carrots, green beans, and cauliflower relative to uncooked vegetables. In their
subsequent research, the scientists achieved comparable results by steaming collard
greens, mustard greens, kale, Brussels sprouts, broccoli, green bell pepper, spinach,
and cabbage [17].
After 4 years, the writers revisited some of the same vegetables from the previ-
ous research, this time using different cooking methods (sautéing, heating, and
steaming). They concluded that sautéing has the highest health potential (binding
bile acids) for mustard greens, kale, broccoli, cabbage, and green bell pepper, while
steaming was the optimal option for collard greens.
It is difficult to forecast the effect and retention of certain vitamins without ade-
quate knowledge about the conditions and methods of food production, transporta-
tion, and handling, emphasising the critical need for further study in this area.
Flavour has been found to be the most critical of the key considerations that con-
sumers consider when choosing food for cooking at home (flavour, texture, nutri-
tion, cost, protection, and convenience). Preparing and cooking legumes and
vegetables to improve their flavour will increase their consumption, especially
among children.
Several experiments have been conducted since the early twentieth century to
explore the effect of preparation and cooking practices on the stability of nutrients
in food. The findings of these studies differ greatly, prompting consumers to wonder
about the best ways to prepare and cook foods so as to preserve their nutritional
qualities, especially in legumes and vegetables. Many other studies have examined
the effects of preparation and cooking on nutritional value and have shown that
legume growth conditions have a substantial effect on nutrient content [37], Table 1
shows protein content in leguminous seeds.
Legumes are high in B-group vitamins including folate, thiamin, and riboflavin
but low in fat soluble vitamins and vitamin C. Folate is an important food that has
been linked to a lower risk of neural tube defects such as spina bifida in newborns.
Legumes are also high in zinc, iron, calcium, selenium, arsenic, copper, potassium,
magnesium, and chromium. These micronutrients play critical physiological roles
in bone health (calcium), enzyme activity and iron metabolism (copper), carbohy-
drate and lipid metabolism (chromium, zinc), protein synthesis, plasma membrane,
stabilisation (zinc). Haemoglobin synthesis iron and antioxidative activity. Legumes
are generally low in sodium, which is beneficial given the recent developments urg-
ing sodium reduction. While legumes have high iron contents, the iron’s bioavail-
ability is low, reducing the quality of legumes as a source of iron. However, as
legumes are eaten with vitamin C-rich foods, iron absorption is enhanced. As a
result, the high iron content will play a significant role in the prevention of anaemia,
especially in reproductive age or pregnant women [11, 42].
There is the largest incidence concentration of iron deficiency and iron defi-
ciency anaemia in low-income countries because of the persistent shortage of vital
nutrients. The WHO promotes iron supplementation policies for families with anae-
mia particularly since these communities do not have access to several bioavailable
sources of iron [45].
The key challenge of supplementation strategy is low rate of execution. In 2006,
a very substantial number of common negative impacts were diagnosed in individu-
als on iron supplementation, which culminated in questions about the protection of
iron supplements. These problems have been made clear by experiments and find-
ings. An expert group from the World Health Organization (WHO) believes that it
is the existence of highly absorbable supplementation, bypassing the developed sys-
tems for safely chaperoning iron and causing an abundance of non-transferrin-
bound iron that undergirds the health hazards associated with iron supplements.
There is a controversy on whether iron distributed in foods is safer to the public than
supplements [45].
Several ways may be used to enhance nutritional iron values. In case of extreme
iron deficiency, nutrients may be given to some communities. In specific situations,
micronutrient pigments can be applied to packaged food products to enhance their
nutritious benefits.
The unhealthy eating habits making more people sick. The main reason behind
people’s sickness is their unhealthy eating habits. About every third citizen is ill
with different diseases. There is a rise in human and livestock disorders. The exis-
tence of multiple illnesses in human race is a topic that crops up in the minds of citi-
zens. The suggestion that doctors have technological instruments to diagnose illness
is insufficient and contested. Currently, physical and mental capacities are in
decline. Man is a social being, but the social ideals are weakening. This begs one the
doubt as to whether such circumstances are happening in our culture and it obvi-
ously is of considerable concern. The World Health Organization (WHO) suggests
that the consumption of fruit and vegetables was less than 20–50%. This is attrib-
uted to customer desires for items that are easy as opposed to those that are safer [50].
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 983
Integrated food is more effective than food that is separated from whole food.
There are supplements for unnatural foods that have been discovered. Carotenoids
play a pivotal role in our well-being as these vitamins are effective in restoring harm
to our body’s liver, cells, and tissue. Likewise, the supplementation of vitamins A
and C will significantly influence radiation treatment and chemotherapy outcomes.
Salmonella resistance is being developed in humans and animals. Nowadays, citi-
zens are looking at alternative remedies like ayurvedic, herbal, and conventional
medications with great anticipation [51].
Iron deficiency is a prevalent problem. Here, the severity of the crisis and the
practical effect this is having on community demand steps to be enforced. The
methods for supporting the importance enhancement in order to address this issue
effectively. This section is an attempt to put food-based practices back in the middle
of the conversation and promote their acceptance at a higher stage [42]. Table 2
shows health benefits of pulses.
Vitamin and mineral shortages arise in both developed and emerging countries.
Primary efforts should be focused on micronutrient deficiency since an approximate
854 million people are undernourished, an approximate 20 million children under
the age of 5 are severely malnourished, and about 1 million children die suffering
from malnutrition per year.
The research study has concentrated primarily on food items, such as drinks, milk,
cereals, and condiments. The fortification of food items with iron means the incor-
poration of iron-containing components to the commodity formula (e.g., meat or its
derivatives). The choice of such components will be informed by a company’s most
valued product attributes, including flavour and colour [14].
Among the food legumes, lentils are an essential food crop that provides protein
as well as micronutrients to both human and animal diets. Legumes are one of the
least expensive forms of protein and micronutrients for vegetarian diets that are
deficient in animal products. The crop is a staple food in certain developed countries
where starvation is prevalent due to Fe deficiency. Biofortification, a genetic solu-
tion, or fortification, a food processing approach, has the potential to improve Fe
concentration and bioavailability. While research towards increasing Fe concentra-
tion and bioavailability by biofortification has begun, there has been little investiga-
tion into genetic strategies for increasing Fe bioavailability in legumes to date [52].
The aim of biofortification is growing the vitamins and other micronutrients in sta-
ple foods. Food fortification is the method of introducing beneficial micronutrients
to foods or drinks at their point of production. Iron fortification is much safer
method of supplying iron than supplement since it often provides many amounts of
servings at once. While iron supplements can increase amounts of body iron faster
than iron therapy, it is less risky.
Many experiments have investigated the efficacy of iron fortification. Despite
iron inclusion to different kinds of food since the early 1940s, its addition to pre-
packaged food still creates technological challenge. The existence of iron induces
chemical disruption in food, triggering organic chemical modifications that some-
times are undesirable to the eater. With the aid of technological challenge, the indus-
try is making use the unsolvable, poorly soluble, or highly chelated iron substances,
but all of them have minimal chemical reactivity. That being said, chemical avail-
ability and solubility do not supply iron absorption. Iron carriers can provide a com-
bination of the assets required to execute this work. These carriers must be adapted
to the corresponding food commodity. This is an illustration that will demonstrate
several possibilities which apply to food. Iron can catalyse reactions with other
ingredients in food [9, 14].
There are other essential factors that affect the bioavailability of added iron.
Though non-haem iron is very well consumed by human body, haem iron is not as
well consumed. Besides the distinct absorption process of non-haem iron and haem
iron, there are interferences between the two. Polyphenols can obstruct the penetra-
tion of haem iron.
Meat-derived additives are found in numerous foods and beverages as fortifi-
cants. This may be a viable solution for certain categories but would not be suitable
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 985
for more widely used products because of the concerns of market approval, pricing,
and supply.
Another type of iron is ferritin, which is a protein used by flora and fauna to
retain extra iron in their bodies. There has been a reported tendency of phytoferritin
as a nutritional fortificants. Folic acid has been available widely but needs to be
synthesised (beans, lentils, etc). Substances that resemble the action of ferritin have
recently demonstrated very high bioavailability. However, they can be challenging
to introduce into certain foods [11, 47].
3.2 Biofortification
4 Disease Management
Nutritional iron deficiency is a major source of anaemia worldwide. This means that
regular diet cannot meet the complete needs of iron in person. Nutritionists agree
that a balanced food community contains adequate nutrients to support health. It is
assumed that if individuals had an adequate supply of healthy food, they might sat-
isfy their dietary requirements. Despite developments in the sectors such as beef
processing and animal husbandry, there are still many instances of meat malnour-
ishment around the world. This suggests that ample food supply does not actually
contribute to appropriate intakes of vitamin and minerals [54]. Factors that induce
iron deficiency anaemia include low wages, overall low food consumption, poor
monotonous diet, and deficiency of micronutrients. A bad diet can lead to nutri-
tional loss and other detrimental consequences, which can be increased by becom-
ing naive about the importance of a diverse diet [55].
Micronutrient-poor food diets are also the major cause of micronutrient deficien-
cies. Due to technical advances, virtually every conventional food variety would be
adequate in order to meet peoples’ dietary requirements. There is need to preserve
and cultivate the right food to maintain balanced diets that support nutritious needs.
The most impacted demographic groups in need of better nutrition are typically
disadvantaged resource-poor peasant farmers and landholding workers who collect
food and fodder from the land and who also have little buying power. Those that is
who are physically weak, like pregnant and lactating mothers, younger kids and
famine-affected communities, who may fail to conform to a diet that is suitable in
quantity or consistency to have acceptable amounts of iron. All deprived and mar-
ginalised communities should get special care.
Frequent turnover of iron contributes to a chronic scarcity. It is one of the major
causes of iron deficiency anaemia in middle- to low-income countries, since they
produce high level of tannins and phytates which decrease the levels of iron.
Therefore, the measures taken to minimise or alleviate these consequences are
described below [6]. There are plenty of potato, fruit, and legume sources that need
to be immediately supported, along with “many leafy vegetables and legumes that
contain significant quantity of iron, with particular focus on growing the consump-
tion of animal commodities that are high in bioavailable iron and in iron absorption
enhancers.” Analysis in India indicated that anaemia was correlated with limited
meat intakes in children under 3 years of age [56].
Adding varied kinds of grains and tubers to a regular cereal or tuber diet will
dramatically improve the nutritional profile. Adding legumes will marginally
increase the iron balance of your diet. In that scenario, bioavailability of that iron
supply is poor. It is not necessary to reach the recommended amounts of iron con-
sumption without any beef, poultry, or fish. Adding 50 grams of meat improves the
person’s iron consumption “as well as the person’s bioavailable iron. The bioavail-
ability of iron (mg/1000 cal)” has been measured for each food portion in four sta-
ple intakes [57]. There should be ample food available to meet the varied human
needs. Many populations across the planet are unable to gain access to a range of
food that is high in micronutrient. A staple-based diet for a male must be varied
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 987
which includes grains, fruits, and some meat. Obviously, this will not be feasible
with those citizens living in deprived neighbourhoods. Another way to protect hun-
ger is by dietary fortification and supplements [58].
The monotonous diets are often likely to be lacking in other essential nutrients
such as minerals and vitamin. People who follow those diets are likely to suffer
from a variety of nutritional deficiencies. By the intake of a larger range of plant
foods, including fruits and vegetables, humans can more easily attain the requisite
vitamins and minerals. A considerable number of “plant-based nutrients or photo-
chemical” can be consumed, and there is continuing research into the positive
impact of these nutrients. Eating a variety of foods has two benefits in terms of miti-
gating what is known as the double burden of malnutrition. Eating a range of kinds
of foods has double advantages in offsetting what is considered double pressure of
malnutrition [59].
High-fat foods have historically not been considered heart healthy; however,
research is increasing on the cardiovascular benefits of nuts as part of a low-fat and
cholesterol-rich diet. Clinical studies, which have directly researched on interaction
between almonds and blood, have shown that people who consume nuts decrease
cholesterol levels by 6–12%. A Life Sciences Research Office study of six clinical
treatments with walnuts reliably showed reductions in total and LDL cholesterol
that could reduce the incidence of cardiovascular disease [60].
In the late 1970s, researchers found that people in some regions of France who
drank red wine had fewer coronary disorders than other Western communities even
though their diet included large amounts of fat. This hypothesis emerged a sequence
of philosophical inquiries into the so-called “French Paradox” which were substan-
tiated by the conclusions of the ensuing studies. By one of the reports, high con-
sumption of one or more alcoholic drinks decreased the risk of heart attack [37, 61].
There is a chance that grape juice may provide advantages comparable to red
wine since both are high in phenolic antioxidants. Grape juice has been shown to
possess the capacity to suppress platelet aggregation [15]. Besides wine and coffee,
another liquid that includes polyphenols is chocolate. Flavonoids from cocoa help
reduce oxidative stress in LDL cholesterol. In a recent clinical study involving 23
participants, chocolate and cocoa powder was found to improve the oxidation of
low-density lipoprotein cholesterol by 8% against subjects eating a typical American
diet [61].
The important factors influencing iron absorption involve the body’s reserves of
iron (often known as the “pharmacodynamic variables”). As well, there are a range
of things often playing an important role in raising or diminishing the bioavailabil-
ity of iron, including haem (the protein inside red meat from cattle, fish, and poultry
blood) and non-haem iron (the newer iron supplements, such as ferrous sulphate
that are obtained from plant sources), the amount of cooking time and temperature,
and the existence of strengthening foods such as meat peptides and vitamin C (as
well as inhibiting foods such as phytic acid and calcium carbonate) [55].
Utilising both of these methods allows the optimal approach to avoid deficien-
cies. To decide the most suitable combination to end up addressing the bad social
nutrition, the condition study should therefore be performed on the extent,
988 S. Jha and B. Jain
5 Conclusion
Worldwide public face various health issues, which can be prevented by enhancing
the quality of nutrients through their diet. Leguminous seeds are rich in proteins,
micronutrients, low-fat content and bioactive compounds includes dietary fibre,
phytosterols, oligosaccharides, and polyphenols. Leguminous seeds are main staple
food in various countries.
Biotechnology: Production of Natural Bioactive Compounds from Leguminous 989
Legumes and vegetables are used every day in our daily life as food. These super
foods are branded as of supreme nutritious supremacy. Legume seeds are a healthy
food for people all over the world since they contain higher quality proteins (around
20–40% protein) than cereals. Aside from protein, it contains sugars, fibre, amino
acids, micronutrients, and a variety of vitamins and minerals [63]. Legumes are also
well known for the presence of various bioactive compounds such as saponins, tan-
nins, flavonoids, Isoflavones, lectins, phytic acid, and so on, which are essential for
its nutraceutical property and provides beneficial effects on human health as well as
aids in the prevention or treatment of certain diseases such as cardiovascular dis-
ease, diabetes, digestive tract diseases, and so on. There are several other underuti-
lised food legume seeds that could be a source of nutraceutical food. The role of
legumes in human diet is projected to grow in the coming future in order to satisfy
the need for protein and other nutrients in the world’s population while also reduc-
ing the risk associated with animal food supply intake. Only little bit of vegetation
information is recorded and distributed, but it can be used without difficulty and
expense. There is also a long way to go for the curative uses of vegetables and only
a limited portion of the natural compounds in these plants have been recorded in the
existing database of medicinal herbs. Due to the advancement of the technology of
medicinal chemistry and pharmacology, there is a need for the identification of
more medicinal ingredients from foods. A great deal of health care providers wants
patients to consume lots of vegetables which are cultivated and grown at home and
for protein intake the best way to consume legume is in roasted form. We are so
naive of the essence and role of fruits and vegetables. It helps designing strategies,
resources, and policies to reduce the threat of food poisoning and promote its natu-
ral health benefits. Plants, including vegetables, as bioreactors for producing vac-
cines should also be explored to full potential for safeguarding human health and
animal welfare.
Iron deficiency anaemia and iron deficiency are also significant public health
issues. The global size and magnitude, together with the disruptive aspect of this
issue, demands immediate intervention to contain this crucial threat. With the
understanding that foods high in iron raise haemoglobin concentration and decrease
anaemia, a lot of attention is being put on iron fortification. This is also because
states, foreign organisations, and donors have found fortification and supplementa-
tion systems to be readily understood and efficient. However, you will find that in
fact this is not possible. Interest has now switched to nutritional methods that have
significant success for minimising anaemia. Health methods concentrate on opti-
mising the nutrition by growing the availability and intake of a sufficient and healthy
set of foods. Food-based interventions play a key function in combating iron and
other micronutrient shortages. There are many viable steps to be taken by foreign
organisations, states, line ministries of agriculture, health, education, industry and
the private sector, societies and households directly that will improve the use and
bioavailability of iron. These methods seek to fix many common nutritional issues
all at once. The simplest approach to solve this issue is food-based interventions.
Consumers have the ability to have healthier food options, wholesome products,
and a healthy farming system.
990 S. Jha and B. Jain
References
23. ILSI (1999) International Life Sciences Institute Safety assessment and potential health ben-
efits of food components based on selected scientific criteria. ILSI North America Technical
Committee on Food Components for Health Promotion. Crit Rev Food Sci Nutr 39:203–316
24. Jeong Ji Hee, Jo, Yu Na, Kim Hyeon Ju, Jin Dong Eun, Kim Dae-Ok, Heo Ho Jin (2014).
Black Soybean Extract Protects Against TMT-Induced Cognitive Defects in Mice. J Med Food
17:83–91
25. Kushi LH, Meyer KA, Jacobs DRJ (1999) Cereals, legumes, and chronic disease risk reduc-
tion: evidence from epidemiologic studies. Am J Clin Nutr 70:451S–458S
26. Lanza E, Hartman TJ, Albert PS, Shields R, Slattery M, Caan B, Paskett E, Iber F, Kikendall
JW, Lance P, Daston C (2006) High dry bean intake and reduced risk of advanced colorectal
adenoma recurrence among participants in the polyp prevention trial. J Nutr 136:1896–1903
27. Lin LZ, Harnly JM, Pastor-Corrales MS, Luthria DL (2008) The polyphenolic profiles of com-
mon bean (Phaseolus vulgaris L.) Food Chem 107:399–410
28. Jukanti AK, Gaur PM, Gowda CLL, Chibbar RN (2012). Nutritional quality and health ben-
efits of chickpea (Cicer arietinum L.): a review. Br J Nutr 108: S11–S26
29. Kalogeropoulos N, Chiou A, Ioannou M, Karathanos VT, Hassapidou M, Andrikopoulos NK
(2010) Nutritional evaluation and bioactive microconstituents (phytosterols, tocopherols,
polyphenols, triterpenic acids) in cooked dry legumes usually consumed in the Mediterranean
countries. Food Chem 121:682–690
30. Bukva M, Kapo D, Huseinbašić N, Gojak-Salimović S, Huremović J (2019) Iron content in
fruits, vegetables, herbs and spices samples marketed in Sarajevo, Bosnia and Herzegovina
Kem Ind 68:281–287
31. Madhujith T, Naczk M, Shahidi F (2004) Antioxidant activity of common beans (Phaseolus
vulgaris L.) J Food Lipids 11:220–233
32. Sayar S, Koksel H, Turhan M (2005) The effects of protein‐rich fraction and defatting on past-
ing behavior of chickpea starch. Starch‐Stärke 57:599–604
33. Ariviani S, Mudalifah I, Ishartani D, Fauza G (2020) Investigation on antioxidant activity,
protein, and whiteness degree of elicited cowpea sprouts flour prepared with various drying
technique. In AIP Conference Proceedings 2219(1):070003
34. Munirasu S, Ramasubramanian V, Arunkumar P. Effect of Probiotics diet on growth and
biochemical performance of freshwater fish Labeo rohita fingerlings. J Entomol Zool 5:
1374–1379
35. Donno D, Cerutti AK, Prgomet I, Mellano MG, Beccaro GL (2015) Foodomics for mulberry
fruit (Morus spp.): analytical fingerprint as antioxidants’ and health properties’ determination
tool. Food Res Int 69:179–188
36. López-Amorós ML, Hernández T, Estrela I (2006) Effect of germination on legume phenolic
compounds and their antioxidant activity. J Food Comp Anal 19:277–283
37. Bourn D and Prescott J (2002) A comparison of the nutritional value, sensory qualities and
food safety of organically and conventionally produced foods. Crit Rev Food Sci Nutr 42:1–34
38. Wickramasinghe S (1988) Nutritional anaemia. Clin Lab 10:117–134
39. Marinangeli CP, Jones PJ (2011). Whole and fractionated yellow pea flours reduce fasting
insulin and insulin resistance in hypercholesterolemia and overweight human subjects. Brit J
Nutr 105:110–117
40. Martı́nez-Navarrete N, Camacho M, Martı́nez-Lahuerta J, Martı́nez-Monzó J, Fito P (2002)
Iron deficiency and iron fortified foods—a review. Food Res Int 35:225–231
41. Salminen S, Bouley C, Boutron-Ruault MC, Cummings J, Franck A, Gibson GR, Isolauri E,
Moreau MC, Roberfroid M, Rowland I (1998) Functional food science and gastrointestinal
physiology and function. Br J Nutr 80:S147–S171
42. Boxall NS, Adak, GK, De Pinna E and Gillespie IA (2011) A Salmonella typhimurium Phage
Type (PT) U320 outbreak in England, 2008: Continuation of a trend involving ready-to-eat
products. Epidemiol Infect 139:1936–1944
43. Bucher HD, Hengstler P, Schindler C, Meiter G (2002). N-3 PUFA in coronary heart disease:
a meta-analysis of randomized controlled trials. Am J Med 112:298–304
992 S. Jha and B. Jain
1 Introduction
There are several techniques which are employed to enhance the solubility of drug
as follows [5]:
1. pH alteration
2. Co-solvency [6]
3. Surface active agents [6]
4. Complexation [7]
5. Micellar solubilization
6. Hydrotropic solubilization
7. Mixed hydrotropic solubilization
1. Each and every weaker solvent can be made a strong solvent for that solute by
proper selection of solubilizers.
2. Solid dispersions of poorly water-insoluble drugs may be developed using com-
bination of proper concentrations (in safe limits) of water-soluble solid excipi-
ents precluding the use of organic solvents (drawbacks of organic solvents
include pollution, high cost, and toxicity due to organic solvents) [8].
3. Mixed solvency can also be employed in titrimetric analysis.
4. Mixed solvency can also be employed in spectrophotometric analysis.
3 Extraction
Extraction is the first step to separate the desired natural products from the raw
materials. Extraction methods include solvent extraction, distillation method, press-
ing, and sublimation according to the extraction principle [13, 14]. Solvent extrac-
tion is the most widely used method. The extraction of natural products progresses
through the following stages:
1. The solvent penetrates into the solid matrix.
2. The solute dissolves in the solvent.
3. The solute is diffused out of the solid matrix.
4. The extracted solutes are collected.
Any factor enhancing the diffusivity and solubility in the above steps will facili-
tate the extraction. The properties of the extraction solvent, the particle size of the
raw materials, the solvent-to-solid ratio, and the extraction temperature.
Novel Eco-Friendly Method of Extraction for Fixed Oils Using Solvent Action of Solid… 995
The selection of the solvent is crucial for solvent extraction. Selectivity, solubil-
ity, cost, and safety should be considered in selection of solvents. Based on the law
of similarity and intermiscibility (like dissolves like), solvents with a polarity value
near to the polarity of the solute are likely to perform better and vice versa [14].
Generally, the finer the particle size is, the better result the extraction achieves.
The extraction efficiency will be enhanced by the small particle size due to the
enhanced penetration of solvents and diffusion of solutes.
The conventional extraction methods include the following:
1. Maceration
2. Percolation
3. Reflux extraction
Some modern extraction methods such as
1. Supercritical fluid extraction
2. Pressurized liquid extraction
3. Microwave-assisted extraction
The evaporation of the organic solvents leads to isolation and concentration of ana-
lytes. Solvents suitable for the extraction should be less polar, highly volatile, and
have low viscosity. Organic solvents with low polarity such as hexane, dichloro-
methane, and diethyl ether are usually chosen as the organic extracting solvent [15].
The pollution and toxicity caused by most of the organic solvent are a big
challenge.
Sesame oil and sesame seeds were purchased from local market. Sesame seeds were
powdered by means of home mixer. All other chemicals and solvents used were of
standard laboratory grade.
Melting point of thymol is 48 °C. One gram of thymol was taken in a test tube and
the test tube was dipped in a hot water bath to melt thymol, a clear colourless liquid
was obtained, 0.1 ml of sesame oil was transferred in test tube, and test tube was
shaken. Sesame oil was dissolved in melted thymol. Again, 0.1 ml of sesame oil was
transferred into the test tube. Same method was repeated. One millilitre of sesame
oil was easily dissolved by 1 g melted thymol (at about 50 °C) [8].
Melting point of menthol is 45 °C. One gram of menthol was taken in a test tube and
the test tube was dipped in hot water bath to melt menthol, when a clear colourless
liquid (melted menthol) was obtained, 0.1 ml of sesame oil was transferred in a test
tube, and test tube was shaken. Sesame oil got dissolved in melted menthol. Again
0.1 ml of sesame oil was transferred in a test tube. Similarly, sesame oil was dis-
solved in melted menthol. One millilitre of sesame oil was dissolved by 1 g of
melted menthol (at about 45 °C). This indicates that even more quantity of sesame
oil may be dissolved in 1 g of melted menthol [8].
One millilitre of above solution was kept in a test tube. Sesame oil (0.1 ml) was
transferred in it and the test tube was shaken. Oil was completely dissolved in solu-
tion. Further, 0.1 ml sesame oil was transferred and the test tube was shaken. Again
oil was dissolved completely. Same procedure was repeated. It was found that 1 ml
solution was able to dissolve 1.4 ml of sesame oil. This study indicated that sesame
oil has very good solubility in 50% w/v solution [8].
Novel Eco-Friendly Method of Extraction for Fixed Oils Using Solvent Action of Solid… 997
One millilitre of above solution was kept in a test tube. Sesame oil (0.1 ml) was
transferred in it, and the test tube was shaken. Oil was completely dissolved in a
solution. Further, 0.1 ml sesame oil was transferred and the test tube was shaken.
Again oil was dissolved completely. Same procedure was repeated. It was found
that 1 ml solution was able to dissolve 0.8 ml of sesame oil. This study indicated that
sesame oil has quite good solubility in mentioned solution [9].
One millilitre of above solution was taken in a test tube. Sesame oil was transferred
in it and the test tube was shaken. Oil was dissolved again. Same procedure was
repeated. It was found that 1 ml of solution was able to dissolve 0.9 ml of sesame
oil. Sesame oil has good solubility in the mentioned solution [9].
Thymol (25 g) and menthol (25 g) were triturated with the help of mortar pestle to
get clear colourless liquid (eutectic liquid). One millilitre of M:T-1:1 was kept in a
test tube and the test tube was shaken. Oil was completely dissolved. Further 0.1 ml
was taken and the test tube was shaken. Again oil was dissolved and the procedure
was repeated. It was found that 1 ml of eutectic liquid was able to dissolve 1 ml of
sesame oil. This study indicated that sesame oil has very good solubility in eutectic
mixture (M:T-1:1) [9].
(a) Thymol—One gram of thymol was kept on weighed stainless-steel plate and
exposed to about 80 °C in oven. It was found that all the thymol was removed
within 45 min.
(b) Menthol—One gram of menthol was kept on weighed stainless-steel plate and
exposed to about 80 °C in hot air oven. It was found that all menthol was
removed within 30 min.
(c) Removability of thymol from an oily solution containing thymol in sesame
oil—weight of a stainless-steel plate was noted. On this plate, an oily solution
containing 1 g thymol and 1 g sesame oil is transferred and again gross weight
998 P. Mulani et al.
is noted. Plate is exposed to about 80 °C in hot air oven. All menthol is removed
within 30 min.
Note: Sesame oil remains on the plate. It does not vaporize at this tempera-
ture (about 80 °C). Thus, we can employ melted thymol for the extraction of
sesame oil from powdered seeds.
(d) Removability of menthol from an oily solution containing menthol in sesame
oil—weight of a stainless-steel plate was noted. On this plate, an oily solution
containing 1 g menthol and 1 g sesame oil is transferred and again gross weight
is noted. Plate is exposed to about 80 °C in an oven. All menthol is removed in
about 30 min.
(e) Removability of thymol or menthol from other oily solutions (using stainless-
steel plate method).
Same procedure (like method C) was used to observe the removability of
thymol and menthol from following oily solutions.
(i) Oily solution containing 1 ml of 50% w/v thymol in ethanolic solution and
1 g sesame oil.
(ii) Oily solution containing 1 ml of 50% w/v menthol in ethanol solution and
0.8 g of sesame oil.
(iii) Oily solution containing 1 ml of eutectic liquid (menthol: thymol 1:1) and
1 g of sesame oil.
(iv) Oily solution containing 1 ml of an ethanolic solution containing 25% w/v
thymol and 25% w/v menthol and 0.9 g of sesame oil.
4.3 Extraction Methods
6 Conclusion
In this work, it is nicely explained that the solubilizing properties of solids can be
employed for extraction of active constituents from various powders of plant mate-
rials giving eco-friendly methods. The use of toxic, pollution causing organic sol-
vents can be minimized. In the present work, the solvent power of ethanol was
improved using solids (menthol and thymol), so that ethanol (a safe class III sol-
vent) could be used to extract sesame oil. Literature survey revealed that hexane
(harmful solvent) is used with ethanol to extract sesame oil (ethanol is a weaker
solvent for sesame oil). Ethanol is a class III organic solvent. Class III organic sol-
vents are better than class II organic solvents (e.g., methanol, chloroform) from the
point of view of safety, pollution, etc.
Sometimes, because of poor solubility of active constituents in ethanol, we are
unable to use ethanol for extraction purpose and we use harmful organic solvents.
Ethanol can be made a strong solvent using menthol, thymol, etc., as reported in
this work.
Extraction with hexane was performed for comparison purpose only. Hexane is
largely employed for extraction of various oils. Also, in some of the extraction
methods, little amount of pulp was found together with extracted oil. This study is
to show that solids can nicely be employed for extraction of active constituent from
the herbal sources. Solids also possess solubilizing properties. Various harmful sol-
vents like hexane, cyclohexane, and chloroform have been employed for the extrac-
tion of various oils. In this study, traces of thymol or menthol left in the extracted oil
are not harmful. During removal of thymol or menthol from extracted portion, thy-
mol and menthol can be collected employing vacuum distillation, so that they can
be recycled. However, distillation shall require exposure to a much higher
temperature.
References
1. Martin A, Bustamanate P, Chun AHC (1994) Physical pharmacy. B. I. Waverly. Pvt Ltd,
New Delhi. pp 103–212. https://pharmabookbank.files.wordpress.com/2019/03/9.3.physical-
pharmacy.pdf
2. Aulton ME (2013) Pharmaceutics “The science of dosage form and design”. Churchill
Livingstone, New Delhi
3. Martin A (2011) “Solubility and distribution phenomena”, Physical pharmacy and pharmaceu-
tical sciences, 6th edn. Lippincott Williams and Wilkins, Philadelphia
4. United States Pharmacopoeia (2000) 24 National Formulary 19. United States Pharmacopoeial
Convention, Inc., pp 2231–2254
5. The United States Pharmacopoeia, USP 30-NF 25, 2007
6. Cooper JW, Gunn C (1985) General pharmacy. CBS Publisher and Distributors, Delhi,
pp 308–333
7. Remington JP (2006) Remington: the science and practice of pharmacy, 21st edn. Lippincott
Williams and Wilkins, Philadelphia, pp 773–774
Novel Eco-Friendly Method of Extraction for Fixed Oils Using Solvent Action of Solid… 1003
1 Introduction
About 50% of plant species have been reported as endemic to the 34 global biodi-
versity hotspots; each hotspot contains 1500 endemic species [1]. The species with
compact populations are not considered to be vulnerable or endangered at present
but they are at risk and scattered within the restricted geographical regions and
moderately more extensive range [2]. Plants act as the best natural purifiers of the
environment and support an essential role in retaining the oxygen cycle, which is
essential for the survival of all forms of life and reducing carbon dioxides in the air
[3]. Ecologically, woody plants support windbreaks and shelterbelts and are used
for the protection of soil erosion, floods, and deserts [4]. The woody tree plants
reduced temperature in the environment through shade and by intercepting, absorb-
ing, and reflecting solar radiation, especially in warmer places [5]. Tree species are
houses of the majority of wild creatures including animals like insects, birds, small
mammals, and reptiles. The woody shrubs and trees on roadsides may protect the
travellers and curves, thus making a natural guide for safe driving. Woody trees
provide timber for the construction of buildings, agricultural implements, boat and
shipbuilding, matches and matchboxes, mathematical instruments, musical
Y. Jeyaram
Department of Botany, PRIST University, Thanjavur, Tamil Nadu, India
P. Prasannan · R. Raju (*)
Department of Biology, The Gandhigram Rural Institute (Deemed to Be University),
Gandhigram, Dindigul, Tamil Nadu, India
A. Pandian
Department of Botany, PRIST University, Thanjavur, Tamil Nadu, India
Division of Research and Innovation, Department of Biotechnology, Saveetha School of
Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
instruments, furniture and cabinetwork, pencil and pen holders, railway carriage
railway sleepers, packing cases and boxes, picture framing, etc. [4]. Moreover,
woody plants play an important role in the ecological balance and forest structure.
In India, people collect the barks, leaves, roots, and sometimes the whole plant
body. About 30% of the entire plant body was used for one-time purposes. In devel-
oped countries such as the United States, they contribute 25% of total drug produc-
tion whereas fast-developing countries such as China and India contribute 80%. The
main reason for decreasing the population rate of woody medicinal plants is the
introduction of alien species and anthropogenic further activities. Hamilton [6] doc-
umented that several hundreds of medicinal plants have been categorized under the
threatened category with extinction risk. Conservation of wild medicinal plants is
difficult through conventional methods such as layering budding, seed germination,
cuttings, and grafting. However, the availability of plant material is not sufficient to
propagate plantlets through conventional methods. To solve this problem, in vitro
propagation can be used and grown successfully.
The plant tissue culture technique is the most efficient technology for large-scale
plant multiplication through micropropagation. In recent years, it is imperative in
the area of plant propagation, secondary metabolites production, pathogen-free
plant production, production of high-yielding plants, and plant improvement.
Endangered and rare plant species have successfully propagated and conserved by
micropropagation. Tissue cultures were employed to preserve plant genetics,
develop more energetic plants, and rapid production of many uniform plants.
Explant surface sterilization was an essential and most perceptive step of plant tis-
sue culture. Surface sterilization has concerned with explants immersed into a suit-
able concentration of chemical sterilant or decontaminators for a particular time of
establishment in contamination-free culture. The axillary and apical bud of
Hildegardia populifolia, an endangered tree, was sterilized by using various steril-
ant at different time duration, Teepol was treated for 5 min and ethanol (70%) for
3 min, by mercuric chloride (0.1%) for 5 min, and finally cleaned with sterilized
distilled water for 4–5 times [7]. Hildegardia populifolia nodal explants were steril-
ized by 1% Bavistin for 30 min and washed with distilled water and 5% teepol for
15 min and mercuric chloride (0.1%) 3 min. The explants were washed using dis-
tilled water before culture [8]. Syzygium densiflorum explants from mother trees
were kept under running tap water (30 min) and Tween-80 (15 min), the earlier to
exterior sterilization. Further, explants were sterilized by mercuric chloride (0.1%)
for 5–10 min, subsequently rinsed with distilled water afterwards by 2% NaOCl for
5–10 min and ethanol (70%) for 5–10 min. The explants were rinsed with sterile
distilled water in a laminar airflow chamber [9]. Explants (leaf and stem) of
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1007
Nothapodytes foetida were washed in running tap water for 10 min and washed with
1–2 drops of Tween-10 followed sterile distilled water (3 times). The explant steril-
ization was attempted to submerge and shack explants in ethanol (70%) for 30 s and
rinsed with sterilized water (2 times). Surface sterilization was carried out with
mercuric chloride (0.1%) for 3 min in addition to sterile distilled water (3 times)
[10]. The in vitro-derived leaves of Zanthoxylum armatum were excised from the
40-day-old shoots by a sterile blade and soaked in a WPM liquid medium contain-
ing different concentrations of TDZ and shaken for different periods of 12, 24, and
36 h followed by cultured on a medium [11]. Shoot tip and nodal explants of
Gaultheria fragrantissima were washed by running tap water (10–15 min) then
treated with Bavistin (2 g−1) for 15–20 min and kept overnight at 25 °C with 70–80%
RH. The explants were again washed with tap water for 20–25 min followed by
4 g−1 Tata Master (15 min). The explants were treated with antibiotics plantomycin
(50 mg−1) and rifampicin (50 mg−1) for 20 min and washed with sterile water.
Further, the explants were sterilized with 0.1% of HgCl2 for 3 min and 100% of
Tween-80 (one to two drops) for 3 min, eventually sterilized in distilled water for
10 min (4–5 times) [12]. The shoot apex and tip explants of Elaeocarpus blascoi
were cleaned by running tap water (30 min) and then immersed in 10% of sodium
hypochlorite (5–8 min) with two or three drops of Teepol over again with sterile
distilled water (3 times). The explants were further treated with ethanol (70%) for
30 s, 0.05% HgCl2 and washed with sterile distilled water or 2–3 times and immersed
in the antimycotic solution for 5 min [13]. Leaf explants of Leptadenia reticulata
were initially treated with 0.1% Bavistin for 10–15 min and 0.1% of mercuric chlo-
ride for 3–4 min and washed by sterile distilled water (6–8 times) in a laminar air-
flow chamber [14]. Leptadenia reticulate nodal and apical shoots were
surface-sterilized by mercuric chloride (0.1%) for 4–5 min and immersed in ethyl
alcohol (90%) for 30–40 s, followed by washing with sterile water (6–7 times).
Then, explants were treated with additives such as adenine sulphate (25 mg−1), argi-
nine and citric acid, and ascorbic acid (50 mg−1) for 10–15 min [15]. Mature and
healthy seeds of Pterocarpus marsupium were sterilized in two stages. The first
stage was washed with running water for 10 min and sterilized by Bavistin (1%) for
5 min followed by rinsed with running tap water (8 min) and imbibed 24 h in dis-
tilled water. In the second step, the seeds were cleaned with HgCl2 (0.1%) for 4 min
and washed with distilled water (4 times) and cultured on nutrient media [16].
Young shoots of Rauvolfia serpentine were washed by using tap water for 30 min
and soaked in labolene (5%) for 5 min. Further, the explants were exteriorly steril-
ized by HgCl2 (0.1%) for 3 min and eventually washed with sterile water 4–5 times
[17]. Plant materials of Atropa acuminata were sterilized with Tween-20 (10 min)
and washed with tap water for 30 min. The nodal and shoot tip explants excised
from shoots were treated with 0.1% of Bavistin (10 min) and surface-sterilized with
HgCl2 (0.1%) for 4 min and rinsed with sterile distilled water 5 time [18]. Shoots of
Syzygium travancoricum were washed with 10% of Nocidet B-300 (wetting agent)
for 5 min and placed running tap water (30 min) and surface disinfected by 70%
ethanol (3 min) followed by HgCl2 (0.2%) for 5 min. Finally, explants were washed
with sterile distilled water (5–6 times) for 10 min [19]. The young leaves and seeds
1008 Y. Jeyaram et al.
of Berberis aristata were washed with tap water and immersed in Tween-20 (0.1%)
and agitated leaves (5–10 min) and seeds (15 min). The explants were treated with
fungicide such as Bavistin, carbendazim for leaves (20 min) and seeds (20 min) and
surface-sterilized by 0.1% of mercuric chloride for leaves (5 min) and seeds (7 min)
and washed with sterilized double-distilled water for (4–6 times) [20]. The healthy
seeds of Sterculia urens were washed with tap water and 5% of Tween-20 was addi-
tionally used with distilled water 3 times. Then, seeds were surface-sterilized with
mercuric chloride (0.5%) for 5–7 min followed by autoclaved distilled water 2–3
times [21]. Explants of Tylophora indica shoots were washed with running tap
water (30 min) and used 5% labolene to soak for 5 min. After that, 0.1% of HgCl2
(3 min) were used for sterilization and finally rinsed with sterile distilled water (4–5
times) [22]. The nodal and shoot tips of Garcinia travancorica were used as explants
and washed thoroughly with tap water for 15 min and washed with liquid detergent
several times. Explants were also sterilized with 1% Clorox for 15 min. Further,
explants were surface-sterilized by HgCl2 (0.01%) for 20 min and subsequently
washed with distilled water. Then, explants were surface-sterilized by ethanol
(70%) for 10 min and also rinsed with sterile distilled water 3 times [23].
5% Tween (10–15 min), 80% ethanol (30 s), and 0.1% HgCl2 (5 min) were
reported to be suitable for sterilizing the axillary bud and node of Ceropegia inter-
media [24]. The cotyledonary explant of Terminalia bellirica was washed with
lanolin soap solution followed by 0.1% HgCl2 for 15 min [25]. But at the same time,
nodal explants of Terminalia bellirica were treated with 20% Bavistin for 5–7 min
and followed by 0.1% HgCl2 for 7 min for in vitro culture [26]. Patel et al. [14]
reported to eradicate the microorganism from the leaf explants of Leptadenia reticu-
lata; it has washed with 0.1% Bavistin (10–15 min) and 0.1% HgCl2 (3–4 min).
Likewise, 0.1% Bavistin (20 min) 0.1% mercuric chloride (4 min) and 70% ethanol
were used for sterilizing the node of Nilgirianthus ciliates [27]. Surface sterilization
of explants Santalum album was effectively done by using Tween-80 (5 min) and
0.075% mercuric chloride (5–6 min) [28].
The nodal explants of Clerodendrum serratum were treated with 1% lanolin and
followed by 0.1% mercuric chloride was reported as effective to sterilize the
explants Couroupita guianensis [29]. Singh et al. [30] studied and recorded that leaf
of Meizotropis pellita was rinsed with two fungicides such as Bavistin 30 min and
0.1% Tween-20 for 5 min. Further, the explants were washed with 70% ethanol
(1 min) followed HgCl2 0.1% for 4 min. The individual concentrations of sodium
hypochlorite and calcium hypochlorite were effective for seed and node sterilization
[31, 32], whereas mercuric chloride has alone used to sterilize Decalepis hamiltonii
[33]. The application of Tween-20, 70% ethanol, and mercuric chloride was used to
eliminate the microorganism from the leaf explant [34]. The collected seeds of
Entada pursaetha were disinfected with Tween-20 (15 min), 70% ethanol, and
finally immersed in 0.1% HgCl2 for 10 min and also with distilled water [35].
Sharmila et al. [36] reported that various sterilization methods have been tested
for eliminating fungal contamination. Among these, Teepol solution (5–10 min),
10% Bavistin, and antibiotics such as ampicillin and rifampicin (15–20 min) fol-
lowed by 70% alcohol (30–60 s) and 0.1% mercuric chloride solution (3–7 min)
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1009
were considered as effective for leaf and stem explants. The leaf explant of Mahonia
leschenaultia was washed with 1% Labolene detergent for 10–20 min and 0.1%
HgCl2 for 5–10 min [37]. Similarly, Sahoo and Chand [38] studied the nodal portion
of Vitex negundo rinsed with 5% Laboline, 7% sodium hypochlorite (7–10 min),
and subsequently washed with 0.1% mercuric chloride for 8 min. Kumar et al. [39]
reported that Caesalpinia bonduc was soaked in sulphuric acid to break the seed
dormancy during the sterilization procedure seeds after that the explants were
washed with mercuric chloride solution.
The seeds of P. santalinus were treated with a combination of 50% HCl + 50
ethyl alcohol for 3 h and air-dried for 2 days before inoculation [40]. The axillary
bud segments, leaf, and nodal explants of Exacum wightianus were washed under
running tap water for 30 min. 0.1% Tween-20 (15 min) followed by 0.5% Bavistin
were used for sterilizing the explants. After subsequent washes with distilled water,
it was treated with 70% ethanol and finally sterilized with (0.1%) HgCl2 [41]. The
pods were soaked with distilled water for 24 h and washed with Tween-20 (15 min)
and followed by savlon antiseptic solution (0.06%). Further sterilization procedures
were carried out with 0.1% HgCl2 (20 min) followed by 70% ethanol (1 min) [42].
De-pulped seeds of Ilex khasiana were washed under running tap water for 10 min
soaked in 5% teepol for 5 min and treated with 70% ethanol (1 min) and finally
surface-sterilized with 15% sodium hypochlorite [43]. Lal and Singh [44] experi-
mented and recorded that the nodal explants of Celastrus paniculatus were washed
with teepol running tap water. Surface sterilization of nodal explants was success-
fully done under aseptic conditions using 0.1% HgCl2 for 3–5 min. Finally, the
nodal region was exposed to absolute alcohol and washed with sterilized distilled
water. The explant of Tylophora indica was soaked in 5% teepol solution for 5 min
followed by freshly prepared HgCl2 (for 3 min) [45, 46]. The nodal segments with
the axillary region of Gymnema sylvestre were surface-sterilized with Tween-20 for
3 min and 0.1% mercuric chloride (8 min) [47]. The seeds of G. sylvestre were dis-
infected with 0.5% HgCl2 containing laboline for 5 min [48]. The successfully ger-
minated 15-day-old seedlings of P. santalinus were collected and soaked in soap
solution to remove the contaminants and soil and the shoots were treated with 0.1%
mercuric chloride solution [49]. The sterilize nodal explants of Celastrus panicula-
tus in HgCl2 containing Tween-20 solutions (5 min) were washed with distilled
water. Further, the nodal explants were disinfected with 0.1% HgCl2 and washed
with double sterile distilled water [50]. Padmalatha and Prasad [51] documented
that the dried pods were soaked in boiling water (100 °C) overnight and treated with
5% sulphuric acid for 10 min. Seeds were further washed with 2% Bavistin for
30 min and nodes were exposed to 15 min by 70% ethanol and 0.1% HgCl2 were
used for disinfecting the pods and seeds.
Celastrus paniculatus nodal and internode explants were washed by running tap
water (30 min) followed by 25% NaOCl (10 min) and consequently washed with
autoclaved distilled water (4–5 times). The explants were further sterilized with
0.1% mercuric chloride (10 min) and washed 4–5 times by autoclaved double-
distilled water and cultured on the medium [52]. The leaf and stem explants of
Rauwolfia tetraphylla washed running tap water (2 min) with Tween-20 (1 min)
1010 Y. Jeyaram et al.
followed by 60% ethanol (2 min) 0.1% mercuric chloride (HgCl2) (2 min) although
the stem and root explants were sterilized with 0.5% Bavistin (10 min), followed by
Tween-20 (2 min) and 0.1% mercuric chloride (4 min) [53]. The pods of Pterocarpus
marsupium were washed by running tap water (15 min) followed by 2% of Teepol
(10 min) and 5% of Tween-20 (4 min) and disinfected with 0.1% mercuric chloride
(HgCl2) for 6 min and cultured on medium [54]. The healthy seeds of Pterocarpus
santalinus were washed with 70% alcohol (1 min) and 0.1% HgCl2, followed by
0.1% sodium dodecyl sulphate (10 min) and washed by double sterile water for 5
times [55].
Plant tissue culture media provide essential components such as macro, micro, vita-
mins, hormones, and carbon source to the plant for their growth and development.
Lavanya et al. [7] reported axillary and apical buds were cultured on MS medium
fortified with different plant growth regulators (PGRs) to induce multiple shoots in
Hildegardia populifolia. Nodal explants inoculated in MS and WPM containing
PGRs were induced shoots [8]. The nodes of Syzygium densiflorum were cultured
on WPM with combinations of PGRs to develop multiple shoots and microshoots
were cultured on half-strength WPM to induce rooting [9]. The leaves and stem
explants of Nothapodytes foetida were induced somatic embryos on MS medium in
addition to PGRs and organic supplements [10]. In vitro raised aseptic leaf explants
of Zanthoxylum armatum were induced calli on WPM with PGRs [11]. Gaultheria
fragrantissima shoots tips and nodal explants cultured on WPM were induced mul-
tiple shoots. The nodal explants induced multiple shoots on MS medium supple-
mented with different concentrations of PGRs in Leptadenia reticulata, Syzygium
travancoricum, Sterculia urens, Tylophora indica, and Garcinia travancorica [15,
19, 21–23]. Syzygium travancoricum developed multiple shoots on both MS and
WPM [19], whereas nodal explants of Elaeocarpus blascoi and Rauvolfia serpen-
tina were induced multiple shoots on WPM [13, 17].
The leaf of Leptadenia reticulata has induced calli on MS medium with various
PGRs and the leaf calli of Leptadenia reticulate transfer to shooting MS medium to
induce multiple shoots [14]. Seeds of Pterocarpus marsupium were induced multi-
ple shoots on MS medium containing suitable PGRs and cotyledonary shoots were
induced roots when transferred to MS medium containing PGRs [16]. The in vitro
raised cotyledonary nodal explants of Pterocarpus marsupium were cultured on MS
medium [56]. The explants of Rhododendron wattii were cultured on WPM-induced
multiple shoots and microshoots were cultured on WPM with suitable rooting PGRs
which induced roots [57]. While microshoots of Rauvolfia serpentina induced root
on WPM [17], explants of shoot tips and nodal of Atropa acuminata were cultured
on MS medium fortified with PGRs and induced shoot proliferation and RT (Revised
tobacco) medium were used for shoot elongation rooting [18]. Leaf-derived callus
of Berberis aristata have induced multiple shoot on WPM [20]. Nodal explants of
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1011
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
1. Hildegardia Axillary and Teepol (5 min), 70% MS BA and KIN Shoot Soil, sand, and 100% Lavanya et al.
populifolia apical bud ethanol (3 min), 0.1% (0.5–3 mg−1) with multiplication vermiculate [7]
(Malvaceae) mercuric chloride (5 min) GA3 (0.1–3 mg−1) (2:1:1) with MS
basal (¼ strength)
Nodal 1% Bavistin (30 min), 5% MS and BA, KIN and TDZ Shoot Garden 98.3% Upadhyay et al.
teepol (15 min), 0.1% WPM (0.5–10 μM) induction soil+manure (3:1) [8]
mercuric chloride (3 min) and soil rite,
vermicompost,
irrigated with MS
basal (¼ strength)
2. Syzygium Nodal Tween-80 (15 min), 0.1% WPM BAP (1.5 mg−1) with Shoot Sand and sterile 58.5% Ramasubbu
densiflorum mercuric chloride (5 min), IBA (1.5 mg−1) multiplication soil (1:1) sprayed and Divya [9]
(Myrtaceae) 2% NaOCl (5–10 min), distilled water
70% ethanol (5–10 min) with
Shoots Sterile distilled water Half- IBA (0.5 mg−1) Rooting micronutrients
strength and
WPM macronutrients
3. Nothapodytes Leaf and stem Washing in liquid detergent MS TDZ (0.5–3.0 mg−1) Somatic – – Khadke and
foetida (1–2 drops of Tween-10 in coconut water (20%) embryos Kuvalekar [10]
(Icacinaceae) distilled water), 70% (v/v) induction
ethanol (30 s), 0.1%
mercuric chloride (3 min)
4. Zanthoxylum Aseptic leaf Soaked in distilled water WPM TDZ (15 μM) and Calli induction Farmyard manure – Purohit et al.
armatum explants (24 h) combination of TDZ and soil (3:1) [11]
(Rutaceae) (6.0 μM) and NAA
(0.5 μM)
5. Gaultheria Shoot tips and 2.0 g−1 Bavistin (15– WPM TDZ (0.22 mg−1) Shoot 1:1:9 farmyard, – Bantawa et al.
fragrantissima nodal 20 min), 0.1% mercuric multiplication sand, and virgin [12]
(Ericaceae) chloride (3 min) with soil
Y. Jeyaram et al.
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
10. Rauvolfia Nodal 5% (v/v) detergent (5 min), WPM BA (5.0 μM) with Multiple shoot Garden soil 50 – Alatar et al.
serpentine 0.1% HgCl2 (3 min) NAA (1.0 μM) induction [17]
(Apocynaceae) Microshoots Sterile distilled water WPM NAA (1.0 μM) Rooting Soil rite 90
Microshoots Sterile distilled water RT medium IBA (1 mg−1) Rooting Vermiculite
11 Syzygium Nodal 70% ethanol (3 min) 0.2% MS and BA (17.7 μM) and Multiple shoot Sterilized sand 40% Anand et al.
travancoricum HgCl2 (5 min) WPM NAA (1.3 μM) induction and soil mixture [19]
(myrtaceae) (1:1)
12 Berberis aristata Young leaves 1% Tween-20 (5–10 min), WPM TDZ (0.5 μM) and Shoot Sterile soil and – Brijwal et al.
(Berberidaceae) (leaf-derived 0.5% Bavistin (20 min), BA (8.88 μM) with multiplication farmyard manure [20]
callus) 0.1% HgCl2 (5 min) NAA (2.68 μM) (3:1)
13. Sterculia urens Nodal 5% Tween-20 (10–15 min), MS TDZ (0.33 mg−1) Shoot Garden soil, 80% Dhiman et al.
(Sterculiaceae) 0.1% HgCl2 (5 min) multiplication organic manure, [21]
coco peat, and
vermiculite
(3:3:2:1)
14. Tylophora indica Nodal 5% detergent, labolene MS BA (2.5 μM), NAA Shoot Sterile garden 100% Faisal et al.
(Asclepiadaceae) (5 min), 0.1% HgCl2 (0.5 μM) and lAA multiplication soil, soil rite, or [22]
(3 min) (100 mg−1) vermiculite
15. Garcinia Nodal Shaking with 1% Clorox MS BAP (4.0 mg−1) Shoot Sterile soil and – Ramasubbu
travancorica (15 min), 0.01% HgCl initiation sand (1:1) et al. [23]
(Clusiaceae) (20 min), 70% ethanol MS BAP (4.0 mg−1) and Multiple
(10 min) NAA (1.0 mg−1) shoots and
shoot
elongation
Y. Jeyaram et al.
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
16. Ceropegia Axillary bud 5% Tween (10–15 min), MS BA (6.66 μM) Multiple Peat moss and – Karuppusamy
intermedia and node 80% ethanol (30 s), 0.1% shoots garden soil(1:1) et al. [24]
(Asclepiadaceae) HgCl2 (5 min)
Petiole and – MS Vermicompost Calli induction – – Kashyap et al.
leaf explants extract and coelomic [62]
fluid in 3:1 ratio
17. Terminalia Cotyledonary Lanolin soap solution 0.1% MS BAP (2.0 mg−1) Shoot Soil rite and – Mehta et al.
bellirica nodal HgCl2 (15 min) BAP (3.5 mg−1) and proliferation sprayed with 1/4 [25]
(Combretaceae) KN (0.5 mg−1) multiple MS salt solution
shoots
Node 20% (v/v) Extran® (Merck, MS ½ MS + BA Shoot Organic manure – Dangi et al.
India) (5–7 min) 0.1% (8.8 μM) + NAA multiplication and garden soil [26]
HgCl2 (Merck, India) (2.6 μM) (1:1)
(7 min) IBA (2.5 μM) Rooting
18. Leptadenia Leaf 0.1% (w/v) Bavistin MS 2,4-D (0.5 mg−1) and Calli Sand, farm yard – Patel et al. [14]
reticulate (10–15 min) and 0.1% (w/v) BAP (0.5 mg−1) proliferation manure, and clay
(Asclepiadaceae) HgCl2 (3–4 min) (3:1:1)
Leaf-derived MS BAP (0.5 mg−1) and Multiple
calli NAA (0.1 mg−1) shoots
Microshoots One-fourth IBA (1.5 mg−1) and Rooting
strength 100 mg−1 Ascorbic
MS acid
19. Sterculia urens Cotyledonary – MS BAP (2.0 mg−1) Multiple Farmyard manure – Purohit and
Roxb. node shoots and soil (1:3) Dave [58]
(Sterculiaceae)
20. Nilgirianthus Nodal 0.1% (w/v) Bavistin MS BA (3 mg−1) and Multiple Vermicompost 100% survival Ramakrishnan
ciliatus (20 min) 0.1% mercuric IAA (0.1 mg−1) shoots and sand, rate et al. [27]
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools
(Acanthaceae) Microshoots chloride (HgCl2) (4 min) Half- IBA (1.0 mg−1) Rooting horticulture soil
and 70% ethanol strength (1:1:1)
MS
1015
(continued)
Table 1 (continued)
1016
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
21. Santalum album Nodal Dipped in a CPT 0.1% MS NAA (0.53 μM), BA Multiple 40%, garden 100% Sanjaya et al.
(Santalaceae) (5 min) Tween-80, [5] (4.44 μM) shoots 50%, sieved sand, [28]
0.075% mercuric chloride and soil 10%)
[5, 6]
Microshoots – Quarter- IBA (98.4 μM) Rooting
strength
MS basal
22. Clerodendrum Nodal Laboline detergent 1% MS BAP (0.5 mg−1) Best shoot bud Sterilized – Upadhyay and
serratum (10 min) 0.1% mercuric induction and vermiculite Koche [59]
(Lamiaceae) chloride (10 min) multiplication
MS NAA (0.5 mg−1) Rooting
23. Couroupita Seeds Bavistin (7–8 min) 0.1% MS IBA (2.0 mg−1) Germination – 86% Shekhawat and
guianensis Nodal mercuric chloride (5–6 min) MS BAP (4.0 mg−1) Multiple Manokari [29]
(Lecythidaceae) alone and shoots
combinations of
BAP and KIN
(1.0 mg−1) + NAA
(0.5 mg−1) with
additives
Microshoots Distilled water ½ MS IBA (2.5 mg−1) Rooting
Y. Jeyaram et al.
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
24. Meizotropis pellita Leaf Fungicide (Bavistin) 30 min MS 2–4, D (9.06 μM) Callus Sand and 55–65% Singh et al.
(Fabaceae) 0.1% Tween-20 (5 min). and 2–4, D induction and farmyard manure [30]
70% ethanol (1 min) and (9.06 μM) + 2-iP proliferation (3:1:1 ratio)
0.1% HgCl2 (4 min) (7.38 μM)
Leaf calli MS BA Shoot
(17.6 μM) + GA3 initiation
(1.0 μM)
Cotyledonary MS KN+ GA3 Multiple
node (4.6 μM + 1.0 μM) shoots
or BA (13.2,
17.6 μM) + GA3
(1.0 μM)
Microshoots MS IBA (4.9 μM) Rooting
25. Commiphora Node (with – MS BAP (2 mg−1) + GA3 Shoot – – Tejovathi et al.
wightii two nodes and (0.5 mg−1) response [60]
(Burseraceae) two
internodes)
26. Pterocarpus Nodal 10% sodium hypochlorite MS Without hormones Shoot Sand soil and 68% Tiwari et al.
marsupium (10 min) induction fame yard (1:1:1) [31]
(Fabaceae) MS IBA (0.2 mg−1) Multiple
shoots
Seeds 2% Teepol (10 min), 5% MS BA (3.0 mg−1) and Shoot Sterilized soil and 74% Tippani et al.
Tween-20 (4 min) and 0.1% IAA (0.5 mg−1) multiplication vermiculite (1:1) [54]
mercuric chloride (6 min)
Microshoots Distilled water ½ MS IBA (3.0 mg−1) Rooting
27. Decalepis Nodal – BA (2.0 mg−1) Shoot Not mentioned 85% Seeni and
arayalpathra proliferation Decruse [61]
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools
(Apocynaceae)
(continued)
1017
Table 1 (continued)
1018
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
28. Calamus Embryo – MS TDZ (0.1 mg−1) Shoot – 90% Seeni and
nagabettai multiplication Decruse [61]
(Arecaceae)
29. Decalepis Buds and 0.1% HgCl2 (5 min) MS BA (1.5 mg1) + IAA Shoot Farmyard – Giridhar et al.
hamiltonii leaves (0.5 mg−1) multiplication manure, red soil [33]
(Apocynaceae) IBA (2.0 0.2 mg−1) Rooting and sand (1:1:1)
30. Citrus indica Seeds 5% Calcium hypochlorite MS TDZ (0.01 mg−1) Calli Mixture of garden – Laskar et al.
(Rutaceae) and NAA multiplication soil, leaf mould [32]
(90.1 mg−1) compost, and
BAP (0.5 mg−1), Shoot river sand (1:2:3)
TDZ (0.25 mg−1) multiplication
and NAA
(0.25 mg−1)
NAA (10 mg−1) Rooting
31. Vitex negundo Leaf 5% Tween-20 (10 min), MS IAA (0.3 mg−1) and Shoot Manure, soil, and Jawahar et al.
(Lamiaceae) 70% ethanol (10–15 s and BAP (0.3 mg−1) induction sand (1:1:1) [34]
0.1% mercuric chloride IBA (0.5 mg−1) Rooting
(2–3 min)
Nodes 5% Laboline, 7% sodium MS BA (2.0 mg−1) + GA3 Shooting Vermicompost 93% Sahoo and
hypochlorite (7–10 min), (0.4 mg−1) Chand [38]
and 0.1% mercuric IBA (1.0 mg−1) Rooting
chloride (8 min)
32. Entada pursaetha Seeds Tween-20 (15 min), 70% MS 5.0 mg−1BAP Shoot Garden soil 70% Vidya et al.
(Mimosae) ethanol and 0.1% HgCl2 organogenesis [35]
(10 min) 5.0 mg−1 NAA Rooting
Y. Jeyaram et al.
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
33. Cayratia pedata Leave and Teepol (5–10 min) 10% MS BAP Calli – – Sharmila et al.
(Vitaceae) stem Bavistin (15–20 min), 70% (0.5 mg−1) + NAA formation [36]
alcohol (30–60 s) 0.1% (0.2 mg−1)
mercuric chloride solution
(3–7 min)
34. Mahonia Leave 1% Labolene detergent MS BA (1.0 mg−1) and Shoot Top soil and river 78.8 and 90.6 Radha et al.
leschenaultii (10–20 min), and 0.1% IAA (0.02 mg−1) formation sand (1:1) [37]
(Berberidaceae) HgCl2 (5–10 min) IBA (1.0 mg−1) Rooting
35. Caesalpinia Seeds Tween-20 (15 min), 70% MS BAP (17.57 μm) and Shooting – – Kumar et al.
bonduc ethanol (5 min), H2SO4 IBA (2.85 μm) [39]
(Ceasalpiniacae) (8 min) and 0.1% HgCl2 IBA (2.95 μm) Rooting
336. Pterocarpus Mesocotyl 50% HCL + 50 ethyl B5 BAP Shooting – – Anuradha and
santalinus explant alcohol (3 h) (3 mg−1) + NAA Pullaiah [40]
(Fabaceae) (1 mg−1)
IAA (0.1 mg−1), Rooting
NAA and IBA
Seeds 70% alcohol (1 min), 0.1% MS NAA + BAP + Kin Shoot Garden soil and 90% Arockiasamy
HgCl2, and 0.1% sodium (0.1 + 1.0+ 1.0 mg−1) multiplication sand (1:1) et al. [55]
dodecyl sulphate (10 min)
Microshoots Distilled water ¼ MS IAA 1.0 mg−1 Rooting
Nodal 2% Bavastin (15 min), 70% MS TDZ (1.0, 2.0 and Shoot Soil rite 20% Padmalatha
ethanol (2 min), and 0.1% 3.0 mg−1) multiplication and Prasad [51]
mercuric chloride (15 min)
Seed 2% Bavistin (30 min), 70% MS KIN (2.0 mg−1) and Shoot
ethanol (2 min) and 0.1% BA (1.0 mg−1) multiplication
mercuric chloride (12 min)
Microshoots Distilled water MS basal – Rooting
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools
(continued)
1019
Table 1 (continued)
1020
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
37. Celastrus Internode 25% NaOCl (10 min), 0.1% MS BA (4.44 μM) Shoot buds Sand and 80% Rao and
paniculatus mercuric chloride (10 min) regeneration farmyard manure Purohit [52]
(Celastraceae) Microshoots Distilled water ¼ MS salts IBA (2.45 mM) Rooting (1:1)
Nodal 0.1% mercuric chloride and MS NAA (0.1 mg−1) and Shoot – – Martin et al.
Tween-20 (5 min) BA (1.5 mg−1) multiplication [50]
Nodal 0.1% mercuric chloride MS BAP (1.0 mg−1) Shoot Soil and sand 70% Lal and Singh
(3–5 min) and absolute multiplication mixture (3:1) [44]
alcohol
Microshoots Distilled water ½ MS NAA (0.5 mg−1) Rooting
38. Rauwolfia Leaf Tween-20 (1 min), 60% MS NAA (5.0 mg−1) Calli induction 2:1 ratio of sterile 86% Rohela et al.
tetraphylla ethanol (2 min), 0.1% garden soil and [53]
(Apocynaceae) mercuric chloride (2 min) sand
Stem and root 0.5% Bavistin (10 min), MS NAA (5.0 mg−1) Calli induction
Tween-20 (2 min), 0.1%
mercuric chloride (4 min)
Leaf and stem Distilled water MS TDZ (0.25 mg−1) Shoot
calluses and BAP (2 mg−1) multiplication
Microshoots Distilled water MS IAA (1.0 mg−1) and Rooting
IBA (1.0 mg−1)
Y. Jeyaram et al.
S. No Name of the plant Explant Types of sterilant used Medium Conc. hormone Response Hardening (%) Restoration Reference
39. Ilex khasiana Seeds 5% Teepol (10 min), 70% MS BA (8.88 μM) and Shoot Black soil: 70% Dang et al. [43]
(Aquifoliaceae) ethanol (1 min), and 15% Kn (4.64 μM) response vermiculite: sand
sodium hypochlorite (2:1:1)
(15 min)
Cotyledonary Distilled water MS
nodal
Leaf Distilled water MS 2,4-D (9.04 μM) and Callus
BA (2.32 μM) initiation
Leaf calli Distilled water MS BA (6.63 μM) Shoot bud
proliferation
Microshoots Distilled water ½ MS IBA (9.84 μM) Rooting
40. Tylophora indica Petiole 5% Teepol (5 min), 0.1% MS 2,4-D (10 μM) and Calli induction Sterile 100% Faisal et al.
(Asclepiadaceae) HgCl2 (3 min) TDZ (2.5 μM) vermiculite [45]
Friable callus Distilled water MS TDZ (2.5 μM) Shoot
multiplication
Microshoots Distilled water ½ MS IBA (0.5 μM) Rooting
Leaf 5% Teepol (5 min), 0.1% MS 2,4,5-T (10 μM) Calli induction Sterile – Faisal and Anis
HgCl (3 min) vermiculite [46]
Leaf calli Distilled water MS Kin (5 μM) Adventitious
shoots
Microshoots Distilled water ½ MS IBA (0.5 μM) Rooting
41. Gymnema Nodal Tween-20 (3 min), 0.1% MS 2,4-D (0.5 mg−1) Calli – – Gopi and
sylvestre R.Br mercuric chloride (8 min) Vatsala [47]
(Apocynaceae) In vitro Distilled water MS BA, KIN and NAA Shoot – – Komalavalli
axillary node (91.0, 0.5 and multiplication and Rao [48]
explants 0.1 mg−1)
Microshoots Distilled water ½ MS IBA (3 mg−1) Rooting
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools
1021
1022 Y. Jeyaram et al.
microshoots, which devoid of hormone [44, 48, 51]. Similarly, the petiole explant of
Tylophora indica showed callus induction and shoot multiplication on MS medium
[22]. The callogenesis of leaf explant of Tylophora indica has shown better response
on MS medium with various PGRs [46]. The cell suspension culture of Gymnema
sylvestre performed better response on MS medium [47]. Martin et al. [50] reported
that nodal explant of C. paniculatus achieved bud breakage and elongation in MS
medium without growth regulator.
Celastrus paniculatus internode explants were cultured on MS medium addition
of PGRs to initiate shoot buds. Microshoots were inoculated on one-fourth MS
medium supplemented with PGRs to induced roots [52]. The leaf, stem, and root of
Rauwolfia tetraphylla were cultured on MS medium addition of PGRs to induced
calli and in vitro-derived leaf and stem calluses were cultured on MS medium with
PGRs to induce multiple shoots although microshoots were cultured on MS medium
helped to develop roots [53]. The healthy seeds of Pterocarpus marsupium and
P. santalinus were inoculated on MS medium to develop multiple shoots and micro-
shoots on one-half and one-fourth MS strength medium to induced shoots and roots,
respectively [54, 55].
Plant growth regulators (PGRs) are natural organic compounds that have stimulated
or inhibited the specific enzymes or enzyme systems inside plant cells and facili-
tated to regulate plant metabolism. The nodal explants of Syzygium densiflorum
were cultured on WPM supplemented with IBA (1.5 mg−1) with BAP (1.5 mg−1) and
obtained 7.7 number of shoots per explants [9]. Nothapodytes foetida leaf and stem
explants induced maximum culture response (90%) of somatic embryo germination
and regeneration on MS medium addition of TDZ (0.5–3.0 mg−1) along with coco-
nut water (20%) [10]. In vitro regenerated leaves of Zanthoxylum armatum were
soaked in distilled water (24 h) and cultured on WPM containing TDZ (15 μM) and
NAA (0.5 μM) in which TDZ (6.0 μM) combination induced 90% of calli from
explants [11]. The shoots tips and nodal explants of Gaultheria fragrantissima
induced 35 shoots per explants from medium fortified with TDZ (0.22 mg−1) [12].
Similarly, TDZ induced multiple shoots in Elaeocarpus blascoi, Pterocarpus mar-
supium, and Sterculia urens [13, 21, 56]. The leaf explants of Leptadenia reticulata
induced calli on medium addition of 2,4-D (0.5 mg−1) and combinations of NAA
(1.0 mg−1) + BAP (0.5 mg−1) induced 48% more compact calli. The leaf-derived
calli developed a greater number of shoots [30] per explants with 8.62 cm of aver-
age length from medium containing BAP (0.5 mg−1) and NAA (0.1 mg−1) [14].
Leptadenia reticulata nodal explants induced [3, 4] multiple shoots on medium con-
taining IAA (0.6 μM) and BA (9 μM) [15]. Seeds of Pterocarpus marsupium were
developed maximum (23.0) number of shoots with 5.14 cm shoot length on medium
fortified with GA3 (0.50 μM) and TDZ (0.50 μM) [16]. The in vitro raised cotyle-
donary nodal explants of Pterocarpus marsupium induced highest [15] number of
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1023
shoots per explants were obtained from medium containing TDZ (0.4 μM) [56]. In
vitro raised Rhododendron wattii nodal explants were developed maximum (7.72)
no. of shoots along with highest shoot length (2.30 cm) from WPM addition of 2iP
(39.36 μM) [57]. Atropa acuminata shoot tips and nodal explants were induced
shoot proliferation 80% with average shoot number (5.8) on medium with IBA
(1 mg−1) and BAP (1 mg−1) [18]. The nodal explants of Syzygium travancoricum
were cultured on both MS and WPM containing combinations of PGRs BA
(17.7 μM) and NAA (1.3 μM) induced 25 shoots per explant [19]. The young leaf-
derived callus of Berberis aristata induced multiple shoots (17.6–26.5) per callus
on medium containing combinations of PGRs TDZ (0.5 μM) and NAA (2.68 μM)
with BA (8.88 μM) [20]. Tylophora indica nodal explants showed a maximum (8.6)
number of shoots with 5.2 cm of average shoot length on medium containing NAA
(0.5 μM), BA (2.5 μM) [22]. The nodal explants of Garcinia travancorica were
inoculated on MS medium fortified with BAP (4.0 mg−1) have obtained 86% of
shoot initiation and combinations of NAA (1.0 mg−1) and BAP (4.0 mg−1) induced
2.8 shoots per node with an almost equal number of shoots (1.6) and shoot elonga-
tion [23]. The half-strength WPM containing IBA induced root on Syzygium densi-
florum and Elaeocarpus blascoi [9, 13] although WPM containing IAA and NAA
induced root on Rhododendron wattii and Rauvolfia serpentina [17, 57]. Microshoots
of Pterocarpus marsupium were induced root on full-strength MS medium with
IBA [16]. Ceropegia intermedia axillary bud and nodal explants have showed maxi-
mum (5.5) shootlets on MS medium containing BA (6.66 μM) [24] although petiole
and leaf explants of Ceropegia intermedia have induced calli on MS medium forti-
fied with PGRs [62]. The cotyledonary node of Terminalia bellirica showed best
shoot proliferation (80%) on MS medium containing BAP (2.0 mg−1), and a higher
number of shootlets [5] were obtained on MS medium containing combinations of
BAP (3.5 mg−1) and KN (0.5 mg−1) [25]. Leptadenia reticulata leaf explants induced
maximum calli on MS medium with BAP (1.0 mg−1) and 2,4-D (0.5 mg−1), respec-
tively. The leaf-derived calli have induced the highest number (30 per explants) of
shoots on BAP (0.5 mg−1) and NAA (0.1 mg−1) and microshoots cultured on one-
fourth strength MS containing IBA (1.5 mg−1) with ascorbic acid (100 mg−1) exhib-
ited (81%) rooting [14]. The cotyledonary nodes of Sterculia urens induced 4.3
shoots on MS medium containing BAP (2.0 mg−1) [58]. Nilgirianthus ciliata nodal
explants induced higher shoots [24] on MS medium containing BA (3 mg−1) and
IAA (0.1 mg−1). The microshoots developed were exhibited 82% of rooting (14
number roots) on half-strength MS medium addition of IBA (1.0 mg−1) [27]. The
nodal explants of Santalum album showed maximum no. of shoots and shoot length
on MS medium with NAA (0.53 μM) and BA (4.44 μM) responded for a maximum
of 50% of roots from microshoots on quarter-strength MS with IBA (1230 μM)
[28]. The nodal explants of Clerodendrum serratum have developed higher shoot
induction and number of shoots with higher shoot length (5.2 cm) on MS medium
containing BAP (0.5 mg−1) and maximum [7] number of root were induced from the
microshoot on MS medium with NAA (0.5 mg−1) [59]. Couroupita guianensis seeds
were germinated (100%) on a medium containing IBA (2.0 mg−1) and nodal explants
induced multiple shoots (4.1 per explants) on a medium containing BAP (4.0 mg−1).
1024 Y. Jeyaram et al.
5 Hardening
Hardening is one of the most important steps in plant tissue culture in which the
micropropagated plants were allowed to grow under controlled environmental con-
ditions. If the plants were immediately transferred to field conditions, they will get
shock from the outside environment. Due to the inconsistency of weather and
humidity, in vitro plantlets get so much stress from the outside environment. There
are so many factors that make it difficult for plants to acclimatize outside
environment.
Seeni and Decruse [61] have standardized the micropropagation protocol of
some of the rare, endangered, and threatened plants (Decalepis arayalpathra and
Calamus nagabettai). After rooting, invitro propagated plantlets were transferred to
pots containing vermiculite and kept in the greenhouse. About 73% of the rooted
plants were reported as survived and transferred to vermiculite-containing planting
medium [59]. The agar adhered from the root of Decalepis hamiltonii were removed
and planted into the soil of the pots, and the humidity was maintained at 60–70%
and allowed to grow for 2 months. The hardening mixture contained 1:1:1 ratio of
farmyard manure, red soil, and sand [33]. The in vitro-developed plantlets of Citrus
indica were hardened with the mixture of garden soil, leaf mould compost and river
sand (1:2:3) [32]. In vitro the rooted Rauvolfia serpentina plantlets were introduced
into different planting substratum with garden soil, soil rite, and vermiculite. After
the 4 weeks of observation, 30–54 plants were reported to survive in soil rite whereas
in vermiculite 51 plants and in garden soil 30 plants were recorded as survived [17].
The in vitro-developed shootlets of Terminalia bellirica were transferred to the
vermiculite-containing polybags [25]. The leaf of in vitro-grown plantlets was
expanded after 3 weeks of observation and transferred into the mist chamber. Patiel
et al. [63] recorded that at 15 °C temperature, the plantlets of Picrorhiza kurroa
were shown 100 ± 0.00a % survival rate on the sand. Where sprayed with Hoagland
solution in a regular 12-day interval, it exhibited better growth. The rooted plantlets
of Commiphora wightii were planted in vermiculite medium wetted with Hoagland’s
solution. On primary hardening, the rooted microshoots were planted into vermicu-
lite and maintained humidity [64].
Before the introduction of in vitro-grown P. marsupium to the soil, it was trans-
ferred to the culture tubes without sucrose. After that, plants were transferred to clay
pots and 70% of the plants were survived with a high degree of uniformity [56].
Similar studies have been done by Ahmad et al. [16] in P. marsupium, in which the
rooted microshoots were transferred to a potted medium containing soil rite. To
maintain the humidity, the microshoots were covered with transparent polybags and
supplemented 16/8 h of photoperiodism. The half-strength MS liquid was watered
for 20 days followed by tape water for alternative days. The acclimatized plants
were slowly transferred to a mixture of soil rite and garden soil containing medium.
The study conducted on the micropropagation of Oroxylum indicum using different
additives was exhibited better shootlets. The rooted plantlets were immersed in dis-
tilled water for 4 days and transplanted to the sand and soil mixtures [65].
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1027
humidity. Seventy-four healthy plantlets were planted into the field and regenerated
plantlets have not shown any morphological variance. The micropropagated plant-
lets of Nilgirianthus ciliates were planted into the campus of Alagappa University
and 100% of the survival rate was obtained [27]. Mao et al. [57] reported success-
fully acclimatized plantlets were reintroduced into the natural habitats and observed
growth of the plant. Sahoo and Chand [38] have also observed after 6 weeks of
observation of plantlets produces flowers and from this experiment reported that, all
the in vitro propagated plantlets of Santalum album showed 100% of survival rate
during field transfer [28]. The nursery-grown plantlets of Mahonia leschenaultia
were planted into Vattakanal shola forests at Palani hills showed 90.6% of survival
rate [37]. Some of the endogenous fungi treated with the root of Sterculia urens
showed high potency of survival rate in field conditions [21].
The fully developed plantlets of Celastrus paniculatus were successfully trans-
ferred to field condition and about 77% of the plantlets were survived [44].
Komalavalli and Rao [48] reported that about 80–85% of the hardened plantlets of
G. sylvestre were survived in field. The ex vitro rooted plantlets were directly intro-
duced in the field condition and showed normal growth [50]. The successfully hard-
ened plantlets of S. densiflorum were planted in the natural habitat of Megamalai
Wildlife Sanctuary and Kodaikanal Wildlife Sanctuary for the better survival rate.
After 2 months of observation, leaf senescence and slow vegetative growth were
observed [9]. At monsoon season, about 100 plantlets of Decalepis arayalpathra
were planted in Kallar and Aryankavu and about 280 plantlets of Calamus nagabet-
tai were reintroduced [61]. Celastrus paniculatus were successfully planted in field
conditions and obtained 80% of survival rate [52]. The in vitro-developed plantlets
of Rauwolfia tetraphylla were transplanted on the field and obtained 86% of sur-
vival rate [53]. The in vitro plantlets of Pterocarpus marsupium were successfully
transferred to the field with 74% survival rate [54]. The in vitro rooted plantlets
were successfully transformed on the field and obtained 90% of survival rate [55].
7 Conclusion
The present review investigates the importance of woody medicinal plants and their
mass propagation. Medicinal plants are prospective sources of therapeutic medi-
cines and have a considerable role in health systems for humans and animals and
maintaining proper health. They are disappearing under exploitation for their
medicinal, ornamental, perfumery uses. However, woody trees timbers are overex-
ploited for construction of the building, agricultural implements, boat and ship
building, musical instruments, and railway sleepers. Therefore, the rare and endan-
gered woody medicinal plants urgent need their conservation. In recent years, in
vitro culture techniques have been envisaged for germplasm conservation to ensure
the survival of endangered plant species and mass multiplication for commercial
and conservation purposes.
1030 Y. Jeyaram et al.
References
18. Ahuja A, Sambyal M, Koul S (2002) In vitro propagation and conservation of Atropa acumi-
nata Royle ex Lindl-an indigenous threatened medicinal plant. J Plant Biochem Biotechnol
11(2):121–124
19. Anand A, Rao CS, Balakrishna P (1999) In vitro propagation of Syzygium travancoricum
Gamble–an endangered tree species. Plant Cell Tissue Organ Cult 56(1):59–63
20. Brijwal L, Pandey A, Tamta S (2015) In vitro propagation of the endangered species Berberis
aristata DC. via leaf-derived callus. In Vitro Cell Dev Biol Plant 51(6):637–647
21. Dhiman M, Sharma L, Singh A et al (2020) Ex situ conservation using in vitro methods of an
endangered plant Sterculia urens Roxb.: a high volume trade plant for Gum Karaya. Ind Crops
Prod 158:113015
22. Faisal M, Ahmad N, Anis M (2007) An efficient micropropagation system for Tylophora
indica: an endangered, medicinally important plant. Plant Biotechnol Rep 1(3):155–161
23. Ramasubbu R, Manikandan G, Sasikala N (2016) In vitro propagation of Garcinia travanco-
rica–an endemic and endangered tree species of Western Ghats, India. Indian J Trop Biodivers
24(1):64–69
24. Karuppusamy S, Kiranmai C, Aruna V et al (2009) In vitro conservation of Ceropegia interme-
dia-an endemic plant of South India. Afr J Biotechnol 8(17):4052–4057
25. Mehta J, Sain M, Mathuriya BL et al (2012) Rapid micropropagation and callus induction of
Terminalia bellirica Roxb.-an endangered plant. Asian J Plant Sci Res 2(3):364–368
26. Dangi B, Khurana-Kaul V, Kothari SL et al (2014) Micropropagation of Terminalia bellirica
from nodal explants of mature tree and assessment of genetic fidelity using ISSR and RAPD
markers. Physiol Mol Biol Plants 20:509–516. https://doi.org/10.1007/s12298-014-0247-1
27. Ramakrishnan R, Largia MJV, Satish L et al (2017) In vitro mass propagation and conservation
of Nilgirianthus ciliatus through nodal explants: a globally endangered, high trade medicinal
plant of Western Ghats. Plant Biosyst Int J Deal All Aspects Plant Biol 151(2):204–211
28. Sanjaya MB, Rathore TS, Rai VR (2006) Micropropagation of an endangered Indian sandal-
wood (Santalum album L.). J For Res 11:203–209. https://doi.org/10.1007/s10310-006-0207-x
29. Shekhawat MS, Manokari M (2016) In vitro propagation, micromorphological studies and
ex vitro rooting of cannon ball tree (Couroupita guianensis aubl.): a multipurpose threatened
species. Physiol Mol Biol Plants 22(1):131–142. https://doi.org/10.1007/s12298-015-0335-x
30. Singh L, Nailwal TK, Tewari L (2013) An in vitro approach for the conservation of Meizotropis
pellita: an endangered and endemic plant. Plant Am J Plant Sci 04:1233–1240. https://doi.
org/10.4236/ajps.2013.46151
31. Tiwari S, Shah P, Singh K (2004) In vitro propagation of Pterocarpus marsupium Roxb.: an
endangered medicinal tree. Indian J Biotechnol 3:422–425
32. Laskar MA, Hynniewta M, Rao CS (2009) In vitro propagation of Citrus indica Tanaka – an
endangered progenitor species. Indian J Biotechnol 8:311–316
33. Giridhar P, Rajasekaran T, Nagarajan S et al (2004) Production of 2-hydroxy-4-
methoxybenzaldehyde in roots of tissue culture raised and acclimatized plants of Decalepis
hamiltonii Wight & Arn., an endangered shrub endemic to Southern India and evaluation of its
performance vis-a-vis plants from natural hab. Indian J Exp Biol 42:106–110
34. Jawahar M, Ravipaul S, Jeyaseelan M (2008) In vitro regeneration of Vitex negundo L. – a
multipurpose woody aromatic medicinal shrub. Plant Tissue Cult Biotechnol 18(1):37–42
35. Vidya SM, Krishna V, Manjunatha BK (2005) Micropropagation of Entada pursaetha DC – an
endangered medicinal plant of Western Ghats. Indian J Biotechnol 4:561–564
36. Sharmila S, Ramya E, Mownika S et al (2020) Micropropagation of an endangered and
endemic medicinal plant Cayratia pedata var. Glabra. Asian J Pharm Clin Res 13(6):191–196
37. Radha RK, Varghese AM, Seeni S (2013) Conservation through in vitro propagation and resto-
ration of Mahonia leschenaultii, an endemic tree of the Western Ghats. Sci Asia 39:219–229.
https://doi.org/10.2306/scienceasia1513-1874.2013.39.219
38. Sahoo Y, Chand PK (1998) Micropropagation of Vitex negundo L., a woody aromatic medici-
nal shrub, through high-frequency axillary shoot proliferation. Plant Cell Rep 18:301–307.
https://doi.org/10.1007/s002990050576
1032 Y. Jeyaram et al.
39. Kumar SR, Krishna V, Pradeepa K et al (2012) Direct and indirect method of plant regenera-
tion from root explants of Caesalpinia bonduc (L.) Roxb.—a threatened medicinal plant of
Western Ghats. Indian. J Exp Biol 50:910–917
40. Anuradha M, Pullaiah T (1999) Propagation studies of red sanders (Pterocarpus santalinus
L.f.) in vitro – an endangered taxon of Andhra Pradesh, India. Taiwania 44:311–324
41. Baluprakash T, Arumugasamy K, Paulsamy S et al (2011) In vitro regeneration of Exacum
wightianum Arn. (Gentianaceae)-an endemic medicinal plant from the Nilgiris, Western
Ghats. J Appl Pharm Sci 1:167–171
42. Chand S, Singh AK (2004) In vitro shoot regeneration from cotyledonary node explants of a
multipurpose leguminous tree, Pterocarpus marsupium Roxb. In Vitro Cell Dev Biol-Plant
40:464–466. https://doi.org/10.1079/IVP2004548
43. Dang JC, Kumaria S, Kumar S et al (2011) Micropropagation of Ilex khasiana, a critically
endangered and endemic holly of Northeast India. AoB Plants 11:1–7. https://doi.org/10.1093/
aobpla/plr012
44. Lal D, Singh N (2010) Mass multiplication of Celastrus paniculatus Willd: an important medic-
inal plant under in vitro conditions via nodal segments. Int J Biodivers Conserv 2:140–145
45. Faisal M, Singh S, Anis M (2005) In vitro regeneration and plant establishment of Tylophora
indica (Burm. F.) Merrill: petiole callus culture. In Vitro Cell Dev Biol Plant 41:511–515.
https://doi.org/10.1079/IVP2005674
46. Faisal M, Anis M (2003) Rapid mass propagation of Tylophora indica Merrill via leaf callus
culture. Plant Cell Tissue Organ Cult 75:125–129. https://doi.org/10.1023/A:1025084203958
47. Gopi C, Vatsala TM (2006) In vitro studies on effects of plant growth regulators on cal-
lus and suspension culture biomass yield from Gymnema sylvestre R.Br. Afr J Biotechnol
5:1215–1219. https://doi.org/10.4314/ajb.v5i12.43029
48. Komalavalli N, Rao MV (2000) In vitro micropropagation of Gymnema sylvestre – a mul-
tipurpose medicinal plant. Plant Cell Tissue Organ Cult 61:97–105. https://doi.org/10.102
3/A:1006421228598
49. Sita GL, Sreenatha KS, Sujata S (1992) Plantlet production from shoot tip cultures of red
sandalwood (Pterocarpus santalinus L.). Curr Sci 62:532–535
50. Martin G, Geetha SP, Raja SS et al (2006) An efficient micropropagation system for
Celastrus paniculatus Willd.: a vulnerable medicinal plant. J For Res 11:461–465. https://doi.
org/10.1007/s10310-006-0237-4
51. Padmalatha K, Prasad MNV (2008) In vitro plant regeneration of Pterocarpus santalinus Lf
(Red Sanders): an endangered medicinal plant and important timber tree. Tree Sci Biotechnol
2(1):1–6
52. Rao MS, Purohit SD (2006) In vitro shoot bud differentiation and plantlet regeneration in
Celastrus paniculatus Willd. Biol Plant 50(4):501–506
53. Rohela GK, Jogam P, Bylla P, Reuben C (2019) Indirect regeneration and assessment of
genetic fidelity of acclimated plantlets by SCoT, ISSR, and RAPD markers in Rauwolfia tetra-
phylla L.: an endangered medicinal plant. Bio Med Res Int 2019:1–14
54. Tippani R, Vemunoori AK, Yarra R et al (2013) Adventitious shoot regeneration from imma-
ture zygotic embryos of Indian Kino tree (Pterocarpus marsupium Roxb.) and genetic
integrity analysis of in vitro derived plants using ISSR markers. Hortic Environ Biotechnol
54(6):531–537
55. Arockiasamy S, Ignacimuthu S, Melchias G (2000) Influence of growth regulators and explant
type on in vitro shoot propagation and rooting of red sandal wood (Pterocarpus santalinus L.).
Indian J Exp Biol 38:1270–1273
56. Husain MK, Anis M, Shahzad A (2007) In vitro propagation of Indian Kino (Pterocarpus
marsupium Roxb.) using thidiazuron. In Vitro Cell Dev Biol Plant 43(1):59–64. https://doi.
org/10.1007/s11627-006-9011-8
57. Mao AA, Vijayan D, Singha RN et al (2018) In vitro propagation of Rhododendron wattii
Cowan—a critically endangered and endemic plant from India. In Vitro Cell Dev Biol Plant
54(1):45–53. https://doi.org/10.1007/s11627-017-9869-7
Conservation Attempts of Woody Medicinal Plants of India by Biotechnological Tools 1033
58. Purohit SD, Dave A (1996) Micropropagation of Sterculia urens Roxb.-an endangered tree
species. Plant Cell Rep 15(9):704–706
59. Upadhyay S, Koche V (2015) Comparison of different medium and establishment of an effi-
cient micropropagation technique of Clerodendrum serratum L. an endangered medicinal
plant. J Environ Sci Toxicol Food Technol 1(1):27–35
60. Tejovathi G, Goswami H, Bhadauria R (2011) In vitro propagation of endangered medicinal
plant-Commiphora wightii. Indian J Sci Technol 4(11):1537–1541
61. Seeni S, Decruse SW (2007) In vitro multiplication and restoration of selected rare, endan-
gered and threatened plants of India. Seed 600:83. In vitro multiplication and restoration of
selected RET plants of India. 3rd Global Botanic Gardens Congress
62. Kashyap S, Kapoor N, Kale RD (2016) Coscinium fenestratum: callus and suspension cell cul-
ture of the endangered medicinal plant using vermicompost extract and coelomic fluid as plant
tissue culture media. Am J Plant Sci 7(06):899–906. https://doi.org/10.4236/ajps.2016.76085
63. Patial V, Sharma M, Bhattacharya A (2017) Potential of thidiazuron in improved micropropa-
gation of Picrorhiza kurroa – an endangered medicinal herb of alpine Himalaya. Plant Biosyst
151:729–736. https://doi.org/10.1080/11263504.2016.1219417
64. Kant T, Tomar UK, Prajapati S et al (2010) In vitro propagation as a viable conservation strat-
egy for Commiphora wightii, an endangered medicinally important desert tree, India. Conserv
Evid 7:94–99
65. Bansal YK, Gokhale M (2012) Effect of Additives on Micropropagation of an Endangered
Medicinal Tree Oroxylum indicum L. Vent. Recent Adv Plant In Vitro Cult 183–196. https://
doi.org/10.5772/50743
66. Patil LM, Murthy HN, Dandin VS et al (2016) Micropropagation of yellow mangosteen: a
valuable endemic tree of India. J For Res 27(1):161–165
67. Gangaprasad A, Decruse SW, Seeni S et al (2005) Micropropagation and ecorestoration of
Decalepis arayalpathra (Joseph & Chandra.) Venter – an endemic and endangered ethnome-
dicinal plant of Western Ghats. Indian J Biotechnol 4:265–270
68. Rathore JS, Rathore V, Shekhawat NS et al (2004) Micropropagation of Woody Plants. Plant
Biotechnology and Molecular Markers. Anamaya Publishers, New Delhi, India, pp 195–205
69. Sharma S, Shahzad A, Ahmad A et al (2014) In vitro propagation and the acclimatization effect
on the synthesis of 2-hydroxy-4-methoxy benzaldehyde in Decalepis hamiltonii Wight and
Arn. Acta Physiol Plant 36:2331–2344. https://doi.org/10.1007/s11738-014-1606-9
70. Dalal NV, Rai RV (2001) In vitro propagation of Ochreinauclea missionis (Wall. EX G. Don),
an ethnomedicinal endemic and threatened tree. In Vitro Cell Dev Biol Plant 37(6):820–823
71. Sharada M, Ahuja A, Kaul MK (2003) Regeneration of plantlets via callus cultures in Celastrus
paniculatus willd,-a rare endangered medicinal plant. J Plant Biochem Biotechnol 12:65–69.
https://doi.org/10.1007/BF03263163
72. Ahmed S (2018) Tissue culture studies on a medicinal plant of Capparidaceae under different
biochemical conditions. IJCRT 6(4):595–598
Conservation of Endangered Medicinal
Plants by In Vitro Propagation Methods
1 Introduction
Medicinal herbs grow naturally all around us, and they have been used to treat ill-
ness and maintain health for centuries by different cultures all over the world. These
easily accessible and culturally significant traditional medicines are a vital source of
livelihood for indigenous and rural populations [1]. For decades, people have used
a wide variety of medicinal plants for their survival; mainly, they have been used in
the primary healthcare system since ancient times. Present-day medications based
on traditional knowledge are developed and preserved by different indigenous com-
munities around the globe. Medicinal plant species have received scientific and
commercial attention since they have immense potential in disease prevention and
curing various diseases. Explorations on plant wealth by various research teams
result in the identification of potential medicinal plants having the efficacy to be
used in the production of plant-based medicines. World Health Organization (WHO)
stated that 80% of developing countries rely on plant-based medicines to treat vari-
ous diseases. Since the usage of medicinal plants is growing rapidly with the
increasing demand for herbal drugs, natural health products, and secondary metabo-
lites [2, 3]. However, these natural resources are declining due to human interven-
tions like habitat destruction, pesticide usage, and overexploitation.
International Union for Conservation of Nature (IUCN) is an international orga-
nization devoted to studying the impact of human interventions on nature and pro-
tecting species and habitats for their survival. The Red List of threatened species
designed by IUCN has become the popular data source on the extinction risk of
species worldwide. Database from IUCN indicates that 50,000–80,000 flowering
plant species are widely used for medicinal purposes. About 15,000 species are
already threatened [4]. Traditional practitioners mainly obtain raw drugs from
Medicinal plants have many advantages; according to one estimate, each new plant-
derived medication is worth $94 million to pharmaceutical corporations and $449
million to society [8]. Since, in recent years, medicinal herbs that exist in nature
have received more attention from the scientific community and in the commercial
trade. According to WHO, 2015 [9], around 60,000 plant species are used medici-
nally worldwide. India is one of the mega biodiversity-rich areas, with nearly 8000
species of medicinal plants belonging to 386 families and 2200 genera of flowering
plants are the primary source of raw drugs [10]. Previous studies signify that most
medicinal plants are flowering plants consisting of 33% trees, followed by herbs,
shrubs, climbers, and lower forms like algae, ferns, and fungi [10]. In recent times,
synthetic drugs have revolutionized health care worldwide, though the majority of
people still depend on herbal medicines to treat various diseases. According to
WHO, traditional medicine is the knowledge, skill, and practices mainly based on
beliefs, theories, and experiences of different indigenous peoples in preventing and
diagnosing physical and mental illness. The most common reasons for dependence
on traditional medicines are more affordable and least side effects than synthetic
medicines [11]. Similarly, the usage of traditional medicines increases when other
practices are ineffective in treating diseases.
Medicinal plants are inevitable since they have been used to prevent, diagnose, and
treat various life-threatening diseases from ancient times. Pharmaceutical industries
and traditional practitioners obtain raw drugs from the leaves, roots, whole plant,
bark, fruits, and seeds that are vital for plant survival and regeneration. However,
Conservation of Endangered Medicinal Plants by In Vitro Propagation Methods 1037
The environmental concern for conserving medicinal plants has become an urgent
focus of discussion. It has been reported that 15,000 species of medicinal plants are
globally threatened due to the loss of habitat, overexploitation, invasive species, and
pollution [4]. The commercial exploitation of medicinal plants for the requirement
of various herbal products is increasing drastically without its restoration. Plants are
described as endangered when their exploitation exceeds 95% and are likely to
become extinct in the near future. The demand for these plant products is expanding
annually at the rate of 15–25% [16]. Therefore, the sustainable use and conservation
of the existing medicinal plant wealth have become a matter of urgency. The in situ
conservation of wild genetic plant diversity in its natural habitat is one of the practi-
cal and appropriate management strategies. Nevertheless, this onsite conservation
will be difficult for the plant species that do not develop viable seeds or too minute
1038 M. V. Lakshmi et al.
Table 1 Details on Indian medicinal plants listed in Red List Database [14]
Si. Vernacular
No. Botanical name name Endemic to Status Uses
1 Aconitum Gaping Himalayan region of CE It is a poisonous plant with
chasmanthum monkshood India and Pakistan over 60 alkaloids,
glycosides, sugars, and
elements in its tuber. Roots
are employed in a variety of
ayurvedic and homeopathic
compositions after they have
been mitigated.
2 Chlorophytum White gold India CE It is effective in alleviating
borivilianum carnal desires and treating
diabetes, arthritis, and natal
and post-natal problems
3 Gentiana Himalayan North Western CE Roots and rhizomes are used
kurroo Gentian Himalaya to treat cough, stomach ache,
and fever.
4 Gymnocladus Minkling Northeast India CE It is used to cure
assamicus dermatological disorders and
to get rid of leaches. The
plant parts are used in
ethno-medicine due to its
anthelminthic properties
5 Lilium White lily Pakistan, CE Underground bulbs are used
polyphyllum Afghanistan, and medicinally as expectorant,
India. astringent, aphrodisiac,
antipyretic, and general
debility
6 Nardostachys Indian nard Mountain range of CE Rhizomes are used to treat
jatamansi Himalaya in Nepal, epilepsy
Bhutan, Myanmar,
Southwest China,
and India
7 Saussurea Indian Jammu Kashmir and CE Roots used as an antiseptic
costus costus Himachal Pradesh in and in treating bronchial
India asthma
8 Tribulus Rajasthan and CE The whole plant is used in
rajasthanensis Gujarat in India treating fever, sterility, and
skin problems
9 Valeriana High altitude region CE Treating the diseases of eye,
leschenaultia of southern-western blood, liver, hysteria,
Ghats hypochondriasis, nervous
unrest, and emotional stress
10 Commiphora Oleo gum Dry regions of CE Used to decrease cholesterol
wightii western India and synthesis in the liver
adjoining areas of
Pakistan
Conservation of Endangered Medicinal Plants by In Vitro Propagation Methods 1039
seeds. The lack of seeds, seed dormancy, non-viable seeds, and endophytic bacterial
and fungal contamination of plant materials are the major limitations in the conven-
tional propagation method of many plants [17].
Totipotency of a cell refers to its ability to develop into a whole organism. In
plant tissue culture system, the totipotent cells are grown in synthetic media, along
with different plant growth regulators (PGR). Previous studies demonstrated the
potential of in vitro culture in raising medicinal plants. The in vitro propagation
techniques are widely used for the production of disease-free plants, genetically
improved varieties, superior genotypes, and plants with enhanced secondary metab-
olite production. Plant tissue culture techniques were established at the end of the
1960s as a probable tool for the worthwhile production of plant secondary metabo-
lites [18]. These micropropagation techniques are highly suitable for endangered,
threatened, and rare species because of their increased coefficient of multiplication
and small demands on the number of initial plants and space [19]. The use of
explants such as apical shoots and axillary buds is better for the conservation of rare
and endangered species [20]. The contamination from different sources like bacteria
and fungi should be strictly avoided for fruitful cultures of plants. The standardiza-
tion of the better combinations of macro- and microelements, vitamins, amino
acids, growth regulators in plant tissue culture studies is the most significant step for
the regeneration of these plants. Many endangered plant species having high-quality
medicinal properties were successfully conserved through different micropropaga-
tion techniques. This is preferable for germplasm conservation to ensure the sur-
vival of endangered plant species, rapid mass propagation for large-scale
re-vegetation, and genetic manipulation studies under precisely controlled physical
and chemical conditions [21]. The integrated approach of these regeneration tech-
niques contributes effective, disease-free, and value-added herbal products for
future industries.
4.1 Seed Culture
Seed culture is an in vitro method for raising sterile seedlings. So far, only a few
medicinal plants have been raised through seed culture. In particular, seeds of
orchids are very minute and hard to germinate in in vivo conditions; hence, seed
culture was found to be an apt method for the regeneration of orchids.
Ethnopharmacological studies conducted in various orchids showed that they are
very significant in treating several diseases such as skin and infectious diseases,
problems concerning the digestive tract, respiratory and reproductive organs, against
tumors, for pain relief, and for lowering fever [22]. Successful establishment of tis-
sue culture protocol for an endangered wild plant species Saussurea esthonica was
achieved from seeds [23]. The optimal conditions for the seed germination and
in vitro seedling growth of Bambara groundnut (Vigna subterranea) based on dif-
ferent culture media compositions were developed by Kone et al. [24]. In vitro
propagation is a proper method to propagate the endangered medicinal orchid,
1040 M. V. Lakshmi et al.
Dendrobium lasianthera through mature seed culture [25]. The nutrient composi-
tion of culture media for orchids is a key factor in seedling growth [26]. Hesami
et al. [27] established a detailed protocol for in vitro seed germination and callus
formation of Ficus religiosa, an important indigenous medicinal woody plant.
Shatnawi et al. [28] successfully multiplied the microshoots derived from the
in vitro germinated seeds of Achillea millefolium and Moringa peregrina in the dif-
ferent concentrations of 6-benzyl amino purine (BA). The ex vitro and in vitro
plants extract of A. millefolium and M. peregrina showed antimicrobial activity
against the selected microorganism.
4.2 Embryo Culture
In embryo culture, embryos from seeds and ovules are allowed to grow in a nutrient-
rich medium. The plantlets develop either directly from the embryo or indirectly
through callus formation. Strychnos potatorum is a medicinally important endan-
gered forest tree effective in treating gonorrhea, leukorrhea, gastropathy, bronchitis,
chronic diarrhea, dysentery, renal and vesicle calculi, diabetes, conjunctivitis, scle-
ritis, and ulcers [29]. Embryo culture was found to be an efficient way to conserve
this medicinally important species using zygotic embryos [30]. Similarly, the
zygotic embryo was used for the in vitro culture of an endangered tree species
Givotia rottleriformis [31] and Kelussia odoratissima [32]. An efficient, reproduc-
ible protocol for the regeneration of Oroxylum indicum, an endangered and medici-
nally important forest tree was also achieved through zygotic embryo culture [33].
In 2022, Kushalan et al. [34] regenerated Curculigo orchioides an endangered
medicinal plant, through somatic embryogenesis. C. orchioides is considered to
have aphrodisiac, immunostimulant, hepatoprotective, antioxidant, anticancer, and
antidiabetic activities [35].
4.3 Callus Culture
4.5 Protoplast Culture
Protoplasts are cells without cell walls, widely used for the regeneration of plantlets.
Protoplast cultures are established by the removal of cell walls either by mechanical
or by enzymatic processes [50]. Protoplast culture and plant regeneration of medici-
nally important plant Tylophora indica were attained through callus regeneration
[51]. Gentiana straminea, an important medicinal plant in Chinese traditional medi-
cine, threatened due to the uncontrolled collection and poor seed germination [52].
Later, Shi et al. [53] developed an effective protocol for the regeneration of G. stra-
minea from its embryonic calli protoplasts.
1042 M. V. Lakshmi et al.
4.6 Bud Culture
Bud culture is a type of plant tissue culture technique widely used for raising plant-
lets either through stem node (single node culture) or axillary buds (axillary bud
culture). Kaur et al. [54] have standardized a quick method for the multiplication
and conservation of Inula racemose, an endangered medicinal plant that is widely
used in traditional medicine in India and Tibet. In addition to this, it is commonly
used in pharmaceutical industries for its antispasmodic, antiasthmatic, and digestive
properties. Dohling et al. [55] established an efficient protocol (axillary bud culture)
for the rapid and large-scale regeneration of Dendrobium longicornu, an endan-
gered and medicinally important epiphytic orchid seriously threaten due to overex-
ploitation and habitat destruction in Northeast India. Nilgirianthus ciliates, a
globally endangered aromatic slender shrub of the Western Ghats with extensive
applications in Ayurveda, was also regenerated through in vitro nodal culture [56].
Lately (2021), Khan et al. [57] developed a well-defined protocol for the large-scale
callus induction of Saussurea costus using four different explants (seeds, leaf, peti-
ole, and internodes) as well as micropropagation from auxiliary buds of S. costus. A
novel method, namely the nylon bag culture system, is considered useful in increas-
ing the growth and development of Jasminanthes tuyetanhiae, a precious medicinal
herb [58]. In order to increase the population of an endangered medicinal plant
Prunus africana, Komakech et al. [59] developed a protocol for micropropagation
using nodal segment explants due to its long flowering cycle and recalcitrant seeds.
P. africana is one of the most popular medicinal plants for treating many diseases,
including prostate cancer, benign prostatic hyperplasia, erectile dysfunction, uri-
nary tract disorders, skin lacerations, kidney disease, chest pain, stomach upset,
inflammation, and as an aphrodisiac.
4.7 Somatic Embryogenesis
5 Conclusion
The wild plant populations from which most medicinal plants are collected are
under threat since they gain more scientific and economic attention around the
globe. Due to human interventions, many medicinal plant species are at the brim of
extinction. Due to commercial exploitation, traditional remedies have occasionally
become unavailable to indigenous peoples who have relied on them for years or
millennia. The study and conservation of medicinal herbs are indispensable since
they have immense potential in curing various life-threatening diseases.
Approximately 15,000 medical plant species are already on the verge of extinction
worldwide. Experts predict that the Earth loses at least one key therapeutic candi-
date every 2 years. Hence, scientists and policymakers are proposing new proce-
dures and policies to safeguard our remaining medicinal treasures in the wild
through various Governmental and non-governmental organizations’ involvement.
References
1. Roberson E (2008) Nature’s pharmacy, our treasure chest: why we must conserve our natural
heritage. Centre for Biological Diversity. www.Biologicaldiversity.Org
2. Piet JH (1952) Logical presentation of the Saiva Siddhanta philosophy. Christian Literature
Society for India, Madras
3. Nalawade SM, Sagare AP, Lee CY, Kao CL, Tsay HS (2003) Studies on tissue culture of
Chinese medicinal plant resources in Taiwan and their sustainable utilization. Bot Bull Acad
Sin 44(2):79–98
4. Pimm S, Russell G, Gittleman J, Brooks T (1995) The future of biodiversity. Science 269:347
5. Raven PH (1987) The scope of plant conservation problem worldwide. In: Bramwell D,
Hamann O, Heywood V, Synge H (eds) Botanical gardens and the world conservation strategy.
Academic, London, pp 19–20
6. Schemske DW, Husband BC, Ruckelshaus MH, Goodwillie C, Parker IM, Bishop JG (1994)
Evaluating approaches to the conservation of rare and endangered plants. Ecology 75:584–606
7. Cole IB, Saxena PK, Murch SJ (2007) Medicinal biotechnology in the genus Scutellaria. In
Vitro Cell Dev Biol-Plant 43(4):318–327
8. Mendelsohn R, Balick MJ (1995) The value of undiscovered pharmaceuticals in tropical for-
ests. Econ Bot 49(2):223–228
9. Romanelli C, Cooper D, Campbell-Lendrum D, Maiero M, Karesh WB, Hunter D, Golden
CD (2015) Connecting global priorities: b iodiversity and human health: a state of knowledge
review. World Health Organistion/Secretariat of the UN Convention on Biological Diversity
1044 M. V. Lakshmi et al.
10. Gopi DK, Mattummal R, Narayana SKK, Parameswaran S (2018) IUCN red listed medicinal
plants of siddha. J Res Siddha Med 1(1):15
11. Benzie IF, Wachtel-Galor S (eds) (2011) Herbal medicine: biomolecular and clinical aspects,
2nd edn. CRC Press/Taylor & Francis, Boca Raton
12. Singh RP, Gupta MK (1990) Soil and vegetation study of Lahaul and Spiti cold desert of
Western Himalayas. Indian Forester 116(10):785–790
13. Fox JL, Nurbu C, Bhatt S, Chandola A (1994) Wildlife conservation and land-use changes in
the Transhimalayan region of Ladakh, India. Mt Res Dev 14:39–60
14. Dhyani A (2015) Critically endangered Indian medicinal plants. Heritage Amruth 11:42–45
15. Singh PR, Singh LJ (2021) In-vitro propagation for improvement of medicinal plants: a review.
J Pharmacogn Phytochem 10(1):1484–1489
16. Kala CP, Dhyani PP, Sajwan BS (2006) Developing the medicinal plants sector in northern
India: challenges and opportunities. J Ethnobiol Ethnomed 2(1):1–15
17. Ramakrishnan R, Periyasamy R, Lakkakula S, Subramani P, Arockiam SR, Manikandan R
(2017) In vitro propagation and conservation of useful endangered medicinal plants with anti-
cancer activity. J Mol Biol Biotechnol 2(3):8
18. Bourgaud F, Gravot A, Milesi S, Gontier E (2001) Production of plant secondary metabolites:
a historical perspective. Plant Sci 161(5):839–851
19. Oseni OM, Pande V, Nailwal TK (2018) A review on plant tissue culture, a technique for
propagation and conservation of endangered plant species. Int J Curr Microbiol App Sci
7(7):3778–3786
20. Chokheli VA, Dmitriev PA, Rajput VD, Bakulin SD, Azarov AS, Varduni TV, Stepanenko VV,
Tarigholizadeh S, Singh RK, Verma KK, Minkina TM (2020) Recent development in micro-
propagation techniques for rare plant species. Plants 9(12):1733
21. Sharma S, Thokchom R (2014) A review on endangered medicinal plants of India and their
conservation. J Crop Weed 10(2):205–218
22. Gutiérrez RM, Orchids P (2010) A review of uses in traditional medicine, its phytochemistry
and pharmacology. J Med Plants Res 4(8):592–638
23. Gailīte A, Kļaviņa D, Ievinsh G (2010) In vitro propagation of an endangered plant Saussurea
esthonica. Environ Expe Biol 8:43–48
24. Kone M, Koné T, Silué N, Soumahoro AB, Kouakou TH (2015) In vitro seeds germination and
seedling growth of Bambara groundnut (Vigna subterranea (L.) Verdc (Fabaceae)). Sci World
J 2015:595073
25. Utami ESW, Hariyanto S, Manuhara YSW (2017) In vitro propagation of the endangered
medicinal orchid, Dendrobium lasianthera through mature seed culture. Asian Pac J Trop
Biomed 7(5):406–410
26. An J, Kim PB, Park HB, Kim S, Park HJ, Lee CW et al (2021) Effects of different growth
media on in vitro seedling development of an endangered orchid species Sedirea japonica.
Plan Theory 10(6):1193
27. Hesami M, Daneshvar MH (2018) Indirect organogenesis through seedling-derived leaf
segments of Ficus religiosa- a multipurpose woody medicinal plant. J Crop Sci Biotechnol
21(2):129–136
28. Shatnawi M, Abubaker S, Odat N, Al-Tawaha AR, Majdalawi M (2021) Antimicrobial activity
and micropropagation of some rare and endangered plants of Jordan. J Ecol Eng 22(6):151
29. Yadav KN, Kadam PV, Patel JA, Patil MJ (2014) Strychnos potatorum: phytochemical and
pharmacological review. Pharmacogn Rev 8(15):61
30. Kagithoju S, Godishala V, Kairamkonda M, Nanna RS (2013) Embryo culture is an efficient
way to conserve a medicinally important endangered forest tree species Strychnos potatorum.
J For Res 24(2):279–283
31. Rambabu M, Upender M, Ujjwala D, Ugandhar T, Praveen M, Rama Swamy N (2006) In vitro
zygotic embryo culture of an endangered forest tree Givotia rottleriformis and factors affecting
its germination and seedling growth. In Vitro Cell Dev Biol-Plant 42(5):418–421
32. Askari-Khorasgani O, Mortazaeinezhad F, Otroshy M, Golparvar AR, Moeini A (2013) Direct
regeneration of an endangered medicinal plant Kelussia odoratissima. Int J Agric Crop Sci
5(17):1969
Conservation of Endangered Medicinal Plants by In Vitro Propagation Methods 1045
33. Talari S, Penchala S, Marka R, Rudroju S, Ramaswamy N (2013) Embryo culture an efficient
tool for conservation of an endangered medicinally important forest tree Oroxylum indicum
Kurz. Int J Biotechnol Res 3:45–52
34. Kushalan S, Yathisha UG, Khyahrii SA, Hegde S (2022) Phytochemical and antioxidant evalu-
ation of in vitro and in vivo propagated plants of Curculigo orchioides. In Vitro Cell Dev
Biol-Plant 11:1–10
35. Chauhan NS, Sharma V, Thakur M, Dixit VK (2010) Curculigo orchioides: the black gold with
numerous health benefits. J Chin Integr Med 8(7):613–623
36. Almemary A (2020) Callus induction and differentiation. Future Agric 3:5–9
37. Benjamin ED, Ishaku GA, Peingurta FA, Afolabi AS (2019) Callus culture for the production
of therapeutic compounds. Am J Plant Biol 4(4):76–84
38. Mitra GC, Chaturvedi HC (1970) Fruiting plants from in vitro grown leaf tissue of Rauvolfia
serpentina Benth. Curr Sci 39:128–129
39. Bakhtiar Z, Mirjalili MH, Sonboli A (2016) In vitro callus induction and micropropagation of
Thymus persicus (Lamiaceae), an endangered medicinal plant. Crop Breed Appl Biotechnol
16(1):48–54
40. Purohit S, Joshi K, Rawat V, Bhatt ID, Nandi SK (2020) Efficient plant regeneration through
callus in Zanthoxylum armatum DC: an endangered medicinal plant of the Indian Himalayan
region. Plant Biosyst Int J Deal All Aspects Plant Biol 154(3):288–294
41. Kumar N, Reddy MP (2011) In vitro plant propagation: a review. J Forest Environ Sci
27(2):61–72
42. Rout GR, Samantaray S, Das P (2000) In vitro manipulation and propagation of medicinal
plants. Biotechnol Adv 18(2):91–120
43. Wala BB, Jasrai YT (2003) Micropropagation of an endangered medicinal plant: Curculigo
orchioides Gaertn. Plant Tissue Culture 13(1):13–19
44. Ismail MF, Abdulla NP, Saleh GB, Ismail M (2010) Anthesis and flower visitors in Curculigo
latifolia Dryand. J Biol Life Sci 1:13–15
45. Kant R (2005) Sweet proteins–potential replacement for artificial low-calorie sweeteners. Nutr
J 4(1):1–6
46. Ali MF, Ratnam W, Heng LY (2007) Genetic effects of arsenic and heavy metals pollutants on
Curculigo latifolia Lumbah. J Biol Sci 7:1155–1162
47. Babaei N, Psyquay Abdullah NA, Saleh G, Lee Abdullah T (2014) An efficient in vitro plant-
let regeneration from shoot tip cultures of Curculigo latifolia, a medicinal plant. Sci World J
2014:275028
48. Kaushal S, Sidana A, Dev K (2014) In-vitro plant production through apical meristem culture
of Gentiana kurroo Royle. J Med Plants Stud 3(1):4–9
49. Pe PP, Naing AH, Soe MT, Kang H, Park KI, Kim CK (2020) Establishment of meristem cul-
ture for virus-free and genetically stable production of the endangered plant Hosta capitata.
Sci Hortic 272:109591
50. Bjowani SS, Rasdan MK (1983) Plant tissue culture: theory and practice. Developments in
crop science, Elsevier Science Publishers, Amsterdam, The Netherlands. 1–373
51. Thomas T, Isolation D (2009) Callus formation and plantlet regeneration from mesophyll pro-
toplasts of Tylophora indica (Burm. F.) Merrill: an important medicinal plant. In Vitro Cell
Dev Biol-Plant 45(5):591–598
52. Cai Y, Liu Y, Liu Z, Zhang F, Xiang F, Xia G (2009) High-frequency embryogenesis and
regeneration of plants with high content of gentiopicroside from the Chinese medicinal plant
Gentiana straminea maxim. In-Vitro Cell Dev Biol-Plant 45(6):730–739
53. Shi G, Yang L, He T (2016) Plant regeneration from protoplasts of Gentiana straminea maxim.
Open Life Sci 11(1):55–60
54. Kaur R, Kashyap A, Majeed S, Chauhan NS (2010) In vitro propagation and conservation of
Inula racemosa Hook. F. an endangered medicinal plant of temperate origin. J Adv Lab Res
Biol 1(1):67–70
1046 M. V. Lakshmi et al.
55. Dohling S, Kumaria S, Tandon P (2012) Multiple shoot induction from axillary bud cultures of
the medicinal orchid, Dendrobium longicornu. AoB Plants 2012:pls032
56. Rameshkumar R, Largia MJ, Satish L, Shilpha J, Ramesh M (2017) In-vitro mass propaga-
tion and conservation of Nilgirianthus ciliatus through nodal explants: a globally endangered,
high trade medicinal plant of Western Ghats. Plant Biosyst Int J Deal All Aspects Plant Biol
151(2):204–211
57. Khan A, Shah AH, Ali N (2021) In-vitro propagation and phytochemical profiling of a highly
medicinal and endemic plant species of the Himalayan region (Saussurea costus). Sci Rep
11(1):1–13
58. Nam NB, Trieu LN, Vu NT, Trung LH, Tra TTH, Tram LT, Dai PH, Tung HT, Nhut DT (2022)
Micropropagation of Jasminanthes tuyetanhiae: an endemic and valuable herb in Vietnam.
Plant Cell Tissue Organ Cult 148(1):35–44
59. Komakech R, Kim YG, Kim WJ, Omujal F, Yang S, Moon BC, Okello D, Rahmat E, Nambatya
Kyeyune G, Matsabisa MG, Kang Y (2020) A micropropagation protocol for the endangered
medicinal tree Prunus africana (Hook f.) Kalkman: genetic fidelity and physiological param-
eter assessment. Front Plant Sci 11:1871
60. Camper ND, Coker PS, Wedge DE, Keese RJ (1997) In-vitro culture of ginkgo. In-Vitro Cell
Dev Biol-Plant 33(2):125–127
61. Jayanthi M, Mandal PK (2001) Plant regeneration through somatic embryogenesis and RAPD
analysis of regenerated plants in Tylophora indica (Burm. f. Merrill.). In-Vitro Cell Dev Biol-
Plant 37(5):576–580
62. Chand S, Sahrawat AK (2002) Somatic embryogenesis and plant regeneration from root seg-
ments of Psoralea corylifolia L., an endangered medicinally important plant. In-Vitro Cell
Dev Biol-Plant 38(1):33–38
63. Tawfik AA, Noga G (2002) Cumin regeneration from seedling derived embryogenic callus in
response to amended kinetin. Plant Cell Tissue Organ Cult 69(1):35–40
64. Joshee N, Biswas BK, Yadav AK (2007) Somatic embryogenesis and plant development in
Centella asiatica L., a highly prized medicinal plant of the tropics. Hortic Sci 42(3):633–637
65. Garro-Monge G, Gatica-Arias AM, Valdez-Melara M (2008) Somatic embryogenesis, plant
regeneration and acemannan detection in aloe (Aloe barbadensis Mill.). Agron Costarric 32:41
66. Chen, S-Y, Chien C-T, Baskin JM, and Baskin CC (2010) Storage behavior and changes in
concentrations of abscisic acid and gibberellins during dormancy break and germination in
seeds of Phellodendron amurense var. wilsonii (Rutaceae). Tree physiology 30(2):275–284
67. Azad MAK, Yokota S, Begum F, Yoshizawa N (2009) Plant regeneration through somatic
embryogenesis of a medicinal plant, Phellodendron amurense Rupr. In-Vitro Cell Dev Biol
Plant 45(4):441–449
68. Baskaran P, Jayabalan N (2009) In-vitro propagation of Psoralea corylifolia L. by somatic
embryogenesis in cell suspension culture. Acta Physiol Plant 31(6):1119–1127
69. Chakraborty A, Bhattacharya D, Ghanta S, Chattopadhyay S (2010) An efficient protocol for
in vitro regeneration of Podophyllum hexandrum, a critically endangered medicinal plant.
Indian J Biotechnol 09(2):217
70. Kumar V, Moyo M, Van Staden J (2017) Somatic embryogenesis in Hypoxis hemerocallidea:
an important African medicinal plant. S Afr J Bot 108:331–336
71. Khajuria AK, Hano C, Bisht NS (2021) Somatic embryogenesis and plant regeneration in
Viola canescens Wall. Ex. Roxb.: an endangered Himalayan herb. Plants 10(4):761
Application of Biotechnology to Produce
Plant-Derived Biologically Important
Compounds
1 Introduction
Secondary metabolites from plants are rich source of bioactive substances that have
a variety of health benefits in both humans and animals. Several distinct phyto-
chemicals can be found in plant foods such as vegetables, fruits, grains, seeds, nuts,
and legumes. Current findings on phytochemical compounds have suggested that
they could be a crucial component of disease-fighting therapeutics and preventa-
tives. These sorts of foodstuffs which include such bioactive constituents are func-
tional foods and, if ingested in a constant and continuous way via meals, would
deliver desired beneficial health effects above their natural features. Furthermore,
individuals can also get fortified nutritional supplements that provide specific bioac-
tive phytoconstituents or a collection of bioactive components. Such nutritional
materials are usually given in higher amounts than it does in a typical diet or even
in a medicinal form in order to improve the health of individuals. Polyphenolic
compounds, terpenes, alkaloids, and saponins are the four more prevalent classes of
bioactive components derived from plants [1]. Based upon the overall optimal activ-
ity, biologically active compounds are generally classed as either primary or sec-
ondary [2]. Trees and shrubs synthesize hundreds of naturally occurring substances
based on physiological phase, tissue distribution (floral and non-floral leaves, fruits,
or bark), and ambient circumstances, including natural or anthropogenic stresses.
Such molecules might play a role in the cell’s fundamental biological mechanisms,
such as regulating cellular development and growth by serving either plant growth
factors, or pheromones [3]. Among those, the much more significant aspects include
polyamines, ethylene, gibberellins, brassinosteroids, cytokinins, abscisic acid, and
auxin. Nonetheless, their primary purpose is ecologically, particularly in terms of
protecting plants from predators, microorganisms, as well as fungus [4]. Plants
defend themselves against pathogenic infection via a variety of signal transduction
pathways, which are dependent upon physiological and biochemical characteristics
(selective or adaptive defences), and proactive modifications generated by infec-
tions (active and inducible defence). The benefits of bioactive compounds are pic-
turized in Fig. 1. Substances is also produced at small levels in response to certain
circumstances, such as terpenes, while transcription could be stimulated to create
excessive proportion in response to structural destruction and pathogenic infection.
Transcription factor compounds comprise phytoalexins, pathogen-related (PR) pro-
teins, glucanases, and chitinases, whereas passively and conventional defensive
chemicals comprise glucans, terpenoids, antimicrobial activity enzymes, antifeed-
ants, and enzyme inhibitors [5]. Glycosylated and concatenated versions of phyto-
chemicals implicated mostly in defensive response enable the plants to synthesize
as well as preserve metabolites in a non-toxic state. For eliminating autoallelopathy
and manufacture active forms fast but only when required, polymerization and par-
ticular distribution (for example, in extracellular environment or even other intracel-
lular organelles) potential options [6]. Across several plants, however, de novo
therapeutic plants:
maintenance of
traditional knowledge, Bioactive Alteration of cellular
novel antibacterial
confirmation of membranes anatomy and
medicinal herbs, compounds pharmaceutical research
physiology
conservation of
biodiversity
In various microorganisms,
halting the creation and
a possible biologically
activities of Nucleic acids
active compound target::
Poor communications in
the Norla cell
cytoplasm components
thrombosis stimulation
From antiquity, herbs were used for therapeutic purposes all across the earth. Plants’
therapeutic properties are determined by their phytochemical constituents, particu-
larly secondary metabolites, that are excellent sources of high-value phytochemical
compounds. Bioactive compounds have complicated chemical characteristics and
thus are created in response to internal or external of stresses in order enable plants
to execute wide range of physiological activities. They are employed in a variety of
sectors, including pharmaceuticals, aesthetics, nutritional supplements, scents,
tastes, and colours. The widespread usage of such compounds in a variety of indus-
tries has prompted researchers to concentrate their efforts on improving synthesis
utilizing plant tissue culture (PTC) technologies and maximizing comparative
advantage in the production utilizing culture system [9]. PTC approaches, which are
not affected by climate or geographic factors, would allow for continuous, scalable,
cost-effective, and practical synthesis of secondary metabolites. The aim of this
systematic review is to evaluate the benefits of employing plant cell culture, the
dispersion of major bioactive molecules in land plants, various strategies involved
in production of secondary metabolites, as well as the industrial value of chosen
naturally occurring substances [10]. Herbs always have contributed immensely in
both clinical practice and research. Plant-based components are used by about 80%
of the worldwide people for essential nutrition and exercise [11, 12]. They are high
in phytonutrients, which provide a magical capacity to treat ailments and can be
employed in a variety of industries, including medicines, aesthetics, and nutritional
supplements. Due to various their cost-effectiveness, convenience, eco-friendliness,
and projected effectiveness equivalent to increased conventional pharmacological
agents, they are receiving greater attention among some of the increasing
1050 N. Iqbal et al.
Table 1 List of biologically active metabolites from plants and their uses
Name of
plant Biologically active metabolites Uses
Datura Tropane alkaloids like hyoscyamine Rheumatoid and arthritis
stramonium scopolamine and atropine discomfort relief, hallucinating
medication, autism and anxiety
treatment, potent mind-altering
drug, anti-inflammatory, and
antibiotic
Species of Ajmaline, α-solanine, ajmalicine, solasodine, Antidote for snake bites,
Rauwolfia serpentine and reserpine, β-carboline, naturally occurring tranquillizer,
reserpiline, alstonine, rescinnamine anxiety, sleeplessness, seizures,
and schizophrenic
Ephedra Proanthocyanidins include eucodelphinidin, Hunger suppressing,
leucopelargonine, leucoanthocyanidin, psychostimulants, asthmatic
lucenine, vicenin-1, and vicenin-2; ephedrine prophylactic, spine anaesthesia-
alkaloids include 1-ephedrine, induced nasal congestion and
d-pseudoephedrine, norephedrine, hypotension, and urine discharge
norpseudoephedrine, methylephedrine, and
methylpseudoephedrine
norephedrine, norpseudoephedrine,
methylephedrine, and
methylpseudoephedrine; and flavonoids
include eucodelphinidin, leucopelargonine,
leucoanthocyanidin, lucenine, vicenin-1, and
vicenin-2
Ginkgo Ginkgo biloba is a type of plant that grows in Anti-inflammatory, antidiabetic,
biloba the United States. antihypertension,
Isorhamnetin, isorhamnetin, quercetin, antihypertension,
kaempferol bilobalide, ginsenoside, antihypertension,
ginkgolide A, B, C, quercetin, bilobalide, antihypertension, and malignancy
ginkgolide A, B, and C
Saussurea Flavonoids, phytosterols, triterpenoids, Anti-inflammatory,
medusa lignans, phenolics, gallic acid, syringin, antithrombotic, anticancer,
chlorogenic acid, ethyl gallate, rutin, immunosuppression,
isoquercitrin, gallic acid, syringin, antispasmodic, antibacterial,
chlorogenic acid, ethyl gallate, rutin, anticonvulsant, and antioxidative
isoquercitrin, and sesquiterpenoids
Thymus Rosmarinic acid, borneol, geraniol, carvacrol, Antibacterial, anti-HIV-1 action,
species thymol, α-terpineol carotenoids antispasmodic, neuroprotective,
antihypertensive,
hypocholesterolemic, strong
antioxidant ability, and flavor
enhancer ingredient
Stevia Essential oils, stevioside, steviol rebaudioside Diabetic treatment, coronary
rebaudiana heart disease, cancers, kidney
disorder, overweight, irritable
bowel syndrome treatment, and
periodontal disease treatment
(continued)
Application of Biotechnology to Produce Plant-Derived Biologically Important… 1051
Table 1 (continued)
Name of
plant Biologically active metabolites Uses
Maclura Isoflavones (scandenone and auriculasin), Cardiac activities, insecticidal,
pomifera prenylated isoflavones (osajin and pomiferin) synthetic dye, abrasive,
antimicrobial, and fungistatic
properties
Citrullus Saponins, steroids, alkaloids, and flavonoids Diabetic treatment, coronary
colocynthis heart disease, cancers, kidney
disorder, overweight, irritable
bowel syndrome treatment, and
periodontal disease treatment
Bacopa Triterpenoid saponins like bacosides Neural relaxant, memory
monnieri enhancer, anticonvulsant,
gastroprotective, free radical
scavenging
Commiphora Monoterpenoids, sesquiterpenoids, Infection, arthritis, rheumatism,
wightii diterpenoids, triterpenoids, steroids, overweight, and lipid metabolic
flavonoids, guggultetrols, and lignans are all abnormalities can all be treated
found in Commiphora wightii
Punica Pelletierine, ellagic acid, phenol, and Anticancer, antiheart illness,
granatum flavonoid are examples of flavonoid antidiabetes, and oral treatment,
compounds immune-modulating properties
Plant secondary metabolites (PSM) have been the subject of discussion in the
research world due to their supposed antiparasitic characteristics. Despite a long
history of indigenous medicinal awareness of the therapeutic and preventive
1052 N. Iqbal et al.
integrity, encompassing water and air, is described as soil health. Mechanical, bio-
chemical, and biological factors of soil are all interconnected. The physical and
chemical properties as well as the soil microbiology are influenced by physical
hierarchy, which in turn determines metabolic processes. Chemical qualities com-
prise cation-exchange capability, pH, saltiness, vitamins and nutrients, as well as
morphology, smoothness, permeability, and bulk density. Plant biodiversity is lost
as a result of agricultural practices that impair plant physiochemical complexity and
ground biodiversity [21]. The benefits of secondary metabolites to plants were
explained in Fig. 2.
Plant secondary
metabolites
involved in
protection against
parasites and fungal
infection
herbivores: Insects,
microbes/viruses: UV- protection and
molasses and competing plants attraction
bacteria and fungi N-storage
vertebrates
polinating insects,
inhibition of seed dispersing
Repellence, toxicity, growth inhibition
germination and animals, root nodule
growth inhibition and toxicity
growth of seedings bacteria and
adapted herbivores
pathways (viz. alkaloid, terpenoid, flavonoids, phenolic, saponin, etc.) had been
engineered using CRISPR/Cas editing through knock-out, knock-in, point muta-
tion, fine-tuning of gene expression, and targeted mutagenesis. This genome-
enhancing tool similarly extends its applicability by incorporating the tools of
artificial and systems biology, useful genomics, and NGS to produce genetically
engineered medicinal plants with advanced trends facilitating the production of
pharmaceuticals and nutraceuticals [24].
To resolve discrepancies in species identification and taxonomy, DNA-based
technologies such as PCR, RFLP, AFLP, RAPD, and genotyping may be used.
Biologically active compounds (alkaloids, flavonoids, terpenoids, etc.) can be pro-
duced in vitro using plant organ and tissue culture procedures under controlled con-
ditions. Recombinant DNA techniques can be utilized to manipulate metabolic
pathways and create protein medicines such as antibodies and hormones.
Bioinformatics and genomics, two new sciences, can be used to identify new drugs
using plant-based materials. Biotechnological strategies, we conclude, can improve
and strengthen medicinal plant research.
1056 N. Iqbal et al.
harmful and must be removed from food, and they act as plant defenders against
insects, diseases, and foragers. The following is a summary of the various goals
achieved/achievable through the use of plant biotechnology: (1) synthesis of advan-
tageous biochemicals (massive-scale cell cultures), (2) through use of biomarkers
and genomic maps to aid traditional breeding efforts, (3) rapid clonal multiplication
(haphazard shoot/bulb/protocorm), (4) generation of haploids to develop homozy-
gous lines quickly (anther culture, ovary culture, interspecific hybridization), (5)
hybrid manufacturing that is hard to procure (embryo rescue, pollination), (6)
removal of the virus (thermotherapy, cryotherapy, or chemotherapy coupled with
meristem culture), (7) plants that reproduce vegetative propagation or that produce
refractory seeds must have their germplasm conserved (cryopreservation and DNA
clones), and (8) vegetational genetic modification (soma clonal variant, somatic
hybridization, and gene transfer).
Advantages of Tissue Cultures in Production of Useful Bioactive Compounds
In vitro propagation has become a dependable approach for mass synthesis of plant
material as the demand for innovative goods generated from plant life has risen.
These and a number of additional advantages of using plant cell culture for huge
quantity of critical bioactive chemicals at the industrial stage provide encourage-
ment for its application. The following are a few of the advantages:
(a) Plant-cultured cells are unaffected by environmental factors.
(b) Cellular cultures gain by synthesizing bioactive secondary metabolites and
going on walks in a controlled setting, regardless of climate or soil conditions.
(c) Production phases can be more geared, as they should be in accordance with
market demand.
(d) By utilizing defined cell strains, a higher level of product production and yield
could be preserved.
(e) Because our understanding of the biosynthesis pathways of popular phyto-
chemicals in plants and cultures is frequently still in its infancy, solutions that
are primarily centred on a cellular and microscopic level were required.
(f) These findings suggest that plant cellular cultures in vitro have the capacity to
produce secondary metabolites in industrial quantities.
(g) Cellular lifestyles will reduce strain on already overharvesting pharmaceutical
and other commercially important plant life. Use of in vitro propagation for the
synthesis of organic compounds and prescription medications has made huge
leaps forward in plant research.
(h) The increased use of herbal products for medical purposes, along with low
product yields and plant harvest supply concerns, has reignited interest in large-
scale plant cellular culture technologies.
(i) The development of more modern molecular biology technologies for produc-
ing transgenic cultures and controlling the translation and regulation of biosyn-
thesis pathway is also likely to be a key step toward creating biological cultures
more widely applicable for secondary metabolite production.
(j) From mutant cellular lines, new synthesis methods may be discovered, poten-
tially leading to the development of novel products [28].
1058 N. Iqbal et al.
Despite more focused efforts and intelligent techniques to enhance value to the
country’s herbal sector, the preferred production has not yet reached its maximum
potential. Despite this, the key challenging scenarios such as a lack of great research
documentation, monographs, uniformity in farming practices (excellent agricultural
practices, gap), good laboratory practice (GLP), and product enhancement and
commercialization persist. Furthermore, increased medicinal herb harvesting raises
concerns about plant extinction and deterioration of medicinal herb habitat, result-
ing in a shortage of plant raw materials, which could stymie efforts to achieve
excessive mass manufacture of secondary metabolites. Biotechnological pro-
grammes, in combination with plant tissue cultures, have been identified as an alter-
nate technique for scaling up secondary metabolite synthesis [29]. This includes
knowledge of naturally occurring substances and their biochemical functions, selec-
tion of the highest-yielding populations, specific genomic expression and regulating
enzyme management, and the use of cost-effective sterile bioreactors. A continuous
effort from the government, academia, and business to further nourish and sustain
the herbal sector is critical, leveraging the country’s wealth of plant species.
Certainly, the characteristics and dynamics of PSM-purposeful microbiome interac-
tions in maximum economic crops are still unknown. A few economically important
plants, such as ginger and garlic, can be used as models to study such interactions
and improve the synthesis of the ideal metabolites allicin and curcumin for com-
mercial purposes. Certainly, understanding the connection between economic crop
PSMs and useful microbiome can lead to stepped-forward agricultural practices that
enhance plant fitness and increase the yield of useful secondary metabolites [30, 31].
References
1. Zhao Y, Wu Y, Wang M (2015) Bioactive substances of plant origin. In: Cheung PCK, Mehta
BM (eds) Handbook of food chemistry. Springer Berlin Heidelberg, Berlin/Heidelberg,
pp 967–1008
2. Sharma M et al (2019) Metabolic engineering strategies for enhancing the production of
bio-active compounds from medicinal plants. In: Natural bio-active compounds. Springer,
Singapore, pp 287–316
3. Depuydt S et al (2016) How plant hormones and their interactions affect cell growth. In:
Molecular cell biology of the growth and differentiation of plant cells. CRC Press/Taylor &
Francis, Boca Raton, p 174
4. Mithöfer A, Maffei ME (2017) General mechanisms of plant defense and plant toxins. In:
Plant toxins. Springer, Dordrecht, pp 3–24
5. Walters D (2011) Plant defense: warding off attack by pathogens, herbivores and parasitic
plants. John Wiley & Sons. New York, United States.
6. Chaves Lobón N, de la Cruz IF, Gallego JCA (2019) Autotoxicity of diterpenes present in
leaves of Cistus ladanifer L. Plants 8(2):27
7. Ghasemzadeh A, Ghasemzadeh N (2011) Flavonoids and phenolic acids: role and biochemical
activity in plants and human. J Med Plant Res 5(31):6697–6703
Application of Biotechnology to Produce Plant-Derived Biologically Important… 1059
8. Giannenas I et al (2013) Essential oils and their applications in animal nutrition. Med Aromat
Plants 2:140. https://doi.org/10.4172/2167-0412.1000140
9. Chandran H et al (2020) Plant tissue culture as a perpetual source for production of industrially
important bioactive compounds. Biotechnol Rep (Amsterdam, Netherlands) 26:e00450
10. Schulz V, Hänsel R, Tyler VE (2001) Rational phytotherapy: a physician’s guide to herbal
medicine. Psychology Press, London
11. Winter JM, Tang Y (2012) Synthetic biological approaches to natural product biosynthesis.
Curr Opin Biotechnol 23(5):736–743
12. Yuan H et al (2016) The traditional medicine and modern medicine from natural products.
Molecules 21(5):559
13. Nasri H et al (2014) New concepts in nutraceuticals as alternative for pharmaceuticals. Int J
Prev Med 5(12):1487
14. Atanasov AG et al (2015) Discovery and resupply of pharmacologically active plant-derived
natural products: a review. Biotechnol Adv 33(8):1582–1614
15. Crozier A et al (2006) Phenols, polyphenols and tannins: an overview. In: Crozier A, Clifford
MN, Ashihara H (eds) Plant secondary metabolites: Occurrence, structure and role in the
human diet. Wiley Online Library, John Wiley & Sons Publisher, Hoboken, New Jersey, pp
1–25. https://doi.org/10.1002/9780470988558.ch1
16. Douglas CJ (1996) Phenylpropanoid metabolism and lignin biosynthesis: from weeds to trees.
Trends Plant Sci 1(6):171–178
17. Eriksen NT (2008) The technology of microalgal culturing. Biotechnol Lett 30(9):1525–1536
18. Fraga CG et al (2010) Basic biochemical mechanisms behind the health benefits of polyphe-
nols. Mol Aspects Med 31(6):435–445
19. Gustafsson C (2009) Tools designed to regulate translational efficiency. In: Smolke C (ed) The
metabolic pathway engineering handbook: tools and applications. Boca Raton, FL, CRC Press,
678. http://dx.doi.org/10.1201/9781420077667.ch9
20. Jacobsen JH, Frigaard N-U (2014) Engineering of photosynthetic mannitol biosynthesis from
CO2 in a cyanobacterium. Metab Eng 21:60–70
21. National Research Council (2010) Toward sustainable agricultural systems in the 21st century.
National Academies Press, Washington, DC
22. Abdallah NA, Prakash CS, AG MH (2015) Genome editing for crop improvement: challenges
and opportunities. GM Crops Food 6(4):183–205. https://doi.org/10.1080/21645698.201
5.1129937. PMID: 26930114; PMCID: PMC5033222
23. Razzaq A, Saleem F, Kanwal M, Mustafa G, Yousaf S, Imran Arshad HM, Hameed MK,
Khan MS, Joyia FA (2019) Modern trends in plant genome editing: an inclusive review of
the CRISPR/Cas9 toolbox. Int J Mol Sci 20(16):4045. https://doi.org/10.3390/ijms20164045.
PMID: 31430902; PMCID: PMC6720679
24. Titanji VP, Ngwa AA, Ngemenya M (2007) Applications of biotechnology techniques to the
study of medicinal plants. Afr J Med Med Sci 36(Suppl):23–29
25. Atsmon J, Brill-Almon E, Nadri-Shay C (2015) Preclinical and first-in-human evaluation of
PRX-105, a PEGylated, plant-derived, recombinant human acetylcholinesterase-R. Toxicol
Appl Pharmacol 287:202–209
26. Karuppusamy S (2009) A review on trends in production of secondary metabolites from higher
plants by in vitro tissue, organ and cell cultures. J Med Plants Res 03:1222–1239
27. Alamgir ANM (2017) Therapeutic use of medicinal plants and their extracts. Pharmacognosy.
In: Rainsford KD (ed) Progress in drug research, vol 73. Springer, Cham, pp 403–426
28. Barker LR, Burton JR, Zieve PD (1995) Principles of ambulatory medicine. Williams &
Wilkins, Baltimore, pp 803–843
29. Schmidt TJ, Khalid SA, Romanha AJ, Alves TM, Biavatti MW, Brun R, Da Costa FB (2012)
The potential of secondary metabolites from plants as drugs or leads against protozoan
neglected diseases. Curr Med Chem 15:2176–2228
30. Pang EL, Loh H-S (2016) Current perspectives on dengue episode in Malaysia. Asian Pac J
Trop Med 9:395–401
31. O’Brien J, Wright GD (2011) An ecological perspective of microbial secondary metabolism.
Curr Opin Biotechnol 22:552–558
Appraisal of Medicinal Plants
for Pharmacological Properties
1 Introduction
Generally, various plant species served as important natural resources in both tradi-
tional and modern medicine all over the world. The medicinal uses of plants and
herbal products have been known for thousands of years [1–3], and medicinal plants
have been playing an important role in the development of human culture since his-
tory as well as its in rural areas of advanced countries have been used as main source
of treatment of diseases [4–6]. In addition, it is considered a rich source of content
that can be used in drug development and synthesis [3, 7, 8]. Furthermore, these
plants are accepted as important food sources [9, 10]. Medicinal plants are used
especially as raw materials in the extraction of active substances used in the synthe-
sis of drugs for the treatment of different types of diseases [8, 11]. Natural products
play a key role as a source of pharmaceutical compounds, and many modern medi-
cines derived from traditional herbal medicines are used in modern pharmacother-
apy nowadays [12]. Furthermore, the phytochemical composition and health
benefits of many known medicinal plant species have not yet been fully investigated
but have a promising future [9, 12, 13]. Terrestrial (crop species and non-crop wild
species) plants provide a wide range of ecosystem support such as food and natural
M. Z. Kocak (*)
Department of Herbal and Animal Production, Vocational School of Technical Sciences, Igdir
University, Igdir, Turkey
e-mail: [email protected]
M. G. Kaysim
Department of Field Crops, Faculty of Agriculture, Igdir University, Igdir, Turkey
drugs. Plants in the agricultural ecosystem can be divided into three distinct groups:
crop plants, non-target terrestrial plants, and non-crop plants in the off-crop area
which are not be affected by any plant conservation product treatments. In addition,
crop species are used as surrogates for non-cultivated non-crop plant species, since
they are easier to cultivate [14–16]. Notable therapeutic medicinal plants are poor
inputs for medicinal crop earnings. Recently, in addition to agroforestry systems,
single crops, intermediates, sequential crops, mixed crops, etc. are grown commer-
cially in different countries [17, 18]. The main disadvantages of cultivation of
medicinal plants include the following: (i) adverse weather conditions like drought,
flood, frost, and acidic heavy rain during germination, growth, and harvesting sea-
sons, (ii) viral and fungal diseases which spread rapidly among closely growing
plants of the same species, (iii) the attack of insects and rodents in the field; (iv)
extreme production cost, and (v) the required environmental conditions for cultiva-
tion of medicinal plants.
Medicinal plants are used to meet health needs and combat various diseases since
humanity first started cultivating them. According to the World Health Organization
(WHO) reports, about 80% of the world’s population rely on medicinal plants for
their health needs [19]. Although all plants in nature are precious, considering their
intended use, medicinal plants have different functions to play in human life. In this
section, we will discuss some of the important medicinal plant families that are
frequently used for medicinal purposes, e.g., Fabaceae, Lamiaceae, Euphorbiaceae,
Apocynaceae, Malvaceae, Apiaceae, and Ranunculaceae families that consist of
many medicinal species [20, 21].
Fabaceae
Fabaceae is a large family of mostly herbaceous plants, including shrubs and trees.
Fabaceae consists of 720–730 genera and 19,500 species. Members of the Fabaceae
are dominant species in nearly every biome from deserts to tropical forests. Fabaceae
is divided into three subfamilies: Mimosoideae, Papilionoidea, and Caesalpinioideae
[22]. They have taproots. There are secondary roots that show veining around the
main roots. They fix the free nitrogen of the air by Rhizobium bacteria. The main
component found in many species of the Fabaceae family is alkaloids [19–21].
Arachis hypogaea, Glycine max, Lens culinaris, Phaseolus spp., and Medicago
sativa are well-known examples of this family [23, 24].
Lamiaceae
Lamiaceae is a family of flowering plants in the form of herbs, rarely trees, and
shrubs. Lamiaceae consist of 236–238 genera and 6500–7170 species. The most
distinctive features of this family of plants are they have a four-cornered body, annu-
lar (verticillate) leaf arrangement along the stem, and essential oils [25]. In addition
to some substances such as cellulose, pectin, and sugar, which are present in
Appraisal of Medicinal Plants for Pharmacological Properties 1063
medicines, there are also essences, the majority of which are formed by essential
oils, and it is known that these substances have important pharmacological effects
[20, 26]. The main component found in many species in Lamiaceae, which is rich in
essential oils, is terpenes [19, 21]. Some considerable members of Lamiaceae
include Lavandula angustifolia, Melissa officinalis L., Mentha arvensis L., Mentha
piperita L., Mentha spicata L., Rosmarinus officinalis L., Salvia officinalis L., and
Thymus vulgaris L. species [27].
Apiaceae
Apiaceae consists mostly of herbs and partly of shrubs and trees. Most Apiaceae are
annual, biennial, or perennial herbs consisting of 428–434 genera and 3500–3780
species [19, 23]. Its leaves are alternately arranged. In most genera, the leaves are
frequently aggregated toward the base. Its inflorescence is usually simple or com-
pound umbels. Umbelliferae, meaning “umbrella-bearers,” was the alternative term
used for Apiaceae because of its flowers that look like umbrellas. The main compo-
nent found in many species of Apiaceae, which is rich in essential oils, is coumarins
[21, 25]. Some prominent members of Apiaceae include Carum carvi L., Coriandrum
sativum L., Foeniculum vulgare, and Pimpinella anisum L. species [20, 27].
Apocynaceae
Apocynaceae is a family of flowering plants that also includes trees, shrubs, and
herbs. Apocynaceae consists of 380–415 genera and 4555–4700 species.
Apocynaceae has five subfamilies: Rauvolfioideae, Apocynoideae, Periplocoideae,
Secamonoideae, Asclepiadoideae [28]. Many of its species are tall trees found in
tropical rainforests, and most are from tropical and subtropical regions. Many of
these plants contain milky sap, and many species are poisonous when eaten [25].
Catharanthus roseus is one of the well-known species. The main component found
in many species in Apocynaceae, which is rich in alkaloids, is cardiac glycosides
[19, 21, 29].
Since the day mankind existed, they collected plant and animal resources from
nature to meet their basic needs such as food, shelter and health. Even today, mil-
lions of people in many developing countries derive some of their income from the
plant and animal sources they collect. Valuable plants such as medicinal plants that
are low in the yield and high cost are still collected or cultivated in developed coun-
tries [30–32]. More than one-tenth of the plant species, about 80,000, are used for
medicinal purposes. 3000 of these plants are traded internationally. However, only
900 of them have been cultured and majority are harvested from the wild [33–35].
Despite such a small number of plant trade, the global medicinal plant export has
reached 2.6 million dollars according to the 2016 data [36]. Although nearly 70% of
1064 M. Z. Kocak and M. G. Kaysim
the medicinal plants used industrially have been cultivated, this ratio is the opposite
as a plant variety. To give an example, about 70% of the plants used for medicinal
purposes in India are found in tropical forest areas and the Himalayan region. While
20% of the known medicinal plants in China are used, only 2% of the medicinal
plants are cultivated. Nearly 150 plant species have been cultivated in Europe [35,
37, 38]. Considering the data, it is assumed that the cultivated medicinal plant is not
more than a few hundred species. However, it should be considered that the data on
medicinal and aromatic plants grown or collected from nature are very limited.
Cultivation is often preferred to protect endemic, rare, or endangered species and
genetic diversity and to combat pests and diseases [39–41]. In addition, it is of
course desirable to obtain a high-quality and high-yielding plant with little effort.
Therefore, considering all these reasons, producers prefer to cultivate the plant or
collect it from the wild, if possible.
Plants have the capacity to synthesize different organic molecules, known as sec-
ondary metabolites. Plant secondary metabolites (PSMs) play a role in plants’ adap-
tation to various environmental constraints and in ensuring the continued existence
of organisms in their ecosystems, but are not needed for organisms to live. In addi-
tion, these compounds differ between the same population of plants in respect of
their amounts and varieties. Secondary metabolites have defense functions against
both biotic and abiotic stresses [67–72]. Nevertheless, steroids and sterols, terpenes
and terpenoids, miscellaneous isoprenoids, volatile oils, resins, phenols and phenyl-
propanoids, glycosides, alkaloids (caffeine, ephedrine, nicotine, cocaine etc.), sapo-
nins, bitter principles, cardioactive compounds, etc., are significant classify of
secondary metabolites of plant origin [73–77]. Plant secondary metabolites (PSMs)
are important protectors against different diseases in plants and used as chemical
defense by plants against both biotic and abiotic stresses. In addition, they are used
as medicines, flavorings, fragrances, and insecticides [71, 78–80]. Ever since the
appearance of human civilization, plants have been one of the significant sources of
medicines. Plant secondary metabolites have been shown to play important roles in
the treatment of cancer and in reducing various in vitro and in vivo tumorigenic and
inflammatory factors [43, 75, 76, 81–83]. Medicinal plants have secondary metabo-
lites such as alkaloids, steroids, glycosides, or other known groups of compounds
which have remarkable pharmaceutical effects such anticancer, antidiabetic, antimi-
crobial, antimalarial, antidysenteric effects, as well as combat various types of dis-
eases [43, 76, 84, 85].
1066 M. Z. Kocak and M. G. Kaysim
Medicinal plants are a rich source of bioactive molecules that have numerous bio-
logical properties. These molecules mostly possess secondary metabolites such as
terpenes, alkaloids, and phenolic compounds of medicinal plants which have phar-
macological effects [86–89]. Furthermore, it is estimated that the plant kingdom
contains more than 200,000 secondary metabolites [46, 86]. The groups of plant
secondary metabolites include the following:
Terpenes
Terpenes are the largest and most diverse group of organic compounds produced
mostly by a variety of class of plant secondary compounds. The name “terpene” is
derived from the word “turpentine,” which in turn comes from the old French ter(e)
bintb, meaning “resin.” They are derived chemically from five-carbon isoprene
units that are assembled in different ways [90–93]. These compound contents in the
plants are mostly well-known for their anticancer effects and biological activities.
Some of the best known terpenes described as having anticancer molecules are car-
vone, carveol, isopulegol, eugenol, citral caryophyllene oxide, limonene, elemene,
and methotrexate, thymol, and carvacrol [91, 94–96]. Moreover, terpenes are abun-
dant in higher plants, conifers, citrus, and eucalyptus and are widely distributed in
the flowers, leaves, stems, and roots of these plants [95, 97, 98].
Alkaloids
Alkaloids are natural bioactive organic compounds that consist of one nitrogen
atom in the heterocyclic ring [99]. In addition, it is one of the largest groups of
amino acid-derived natural low-molecular-weight products [11] or all alkaloids
existing in nature [100]. Alkaloids can be discrete according to its fundamental
chemical structure into different types. The basic types of alkaloids are aromatics,
ephedras, carbolines, acridones, indoles, indolizidines, oxindoles, purines, pyrro-
loindoles, phenylethylamines, bisindoles, ergots, imidazoles, quinazolines, quino-
lines, piperidines, pyrrolizidines, manzamines, phenylisoquinolines, pyridines,
pyrrolidines, and simple tetrahydroisoquinolines [59, 101–103]. Apart from their
anticancer properties, these compounds have been found to have many important
bioactivities (such as anti-angiogenesis, autophagy, apoptosis, differentiation, cell
cycle stop, resistant chemoprevention, and anti-metastasis) in human and animal
health. In addition, alkaloids play an important role in increasing the susceptibility
of multidrug resistance (MDR) in human cancer cell lines [80, 91, 94, 104, 105].
Phenolics
Phenolics probably constitute the largest group of plant secondary metabolites
widely distributed in nature [91, 106]. These phenolics are pharmacologically valu-
able for to their anti-inflammatory activities such as quercetin or antihepatotoxic
properties such as silybin. According to structures or biosynthetic origin, phenolics
can be classified into tannins, coumarins, lignans, simple phenolics, flavonoids, stil-
benes, chromones, and xanthones [107, 108]. Phenolics and flavonoids, as are many
other important bioactive agents, have gained widespread interest because of their
Appraisal of Medicinal Plants for Pharmacological Properties 1067
1.7 Pharmacology
Many medicinal plants have long been confirmed as significant sources of therapeu-
tically active compounds. Herewith many research showed that a wide range of
medicinal plants exerted antioxidant, antimicrobial, analgesic, and anti-inflammatory
activities [116–118]. Also, medicinal plants and their parts (leaves, roots, flowers,
seeds, and rhizomes) are used as fresh or dried, decoctions in powder form, tea, and
different extracts (water or alcohol extracts, juices, essential oils, tinctures, balsams,
resins) and used as part of medical treatments [118, 119].
Antioxidant
Antioxidants are free radical scavengers that help in delaying or preventing oxida-
tion. The vital concentration of free radicals or reactive oxygen species (ROS) in
living organisms is maintained by enzymatic antioxidants such as glutathione super-
oxide dismutase, peroxidase, catalase, glutathione reductase, and nonenzymatic
antioxidants such as glutathione, alpha-tocopherol, and ascorbic acid. Also, shown
that fruits rich in polyphenols exert both antioxidant [120, 121]. Many epidemio-
logical research have indicated a reverse relationship between antioxidants from
plant products and the ratio of diseases. Because medicinal plant antioxidants have
removed the free radicals [118, 122, 123], medicinal plants and their materials can
be important promising sources of antioxidants. Furthermore, many of its compo-
nents may contribute to its antioxidant and other protective properties. Among them
flavonoids, phenolic compounds, and vitamins show effective antioxidant activities
[124–127].
Antimicrobial
Medicinal plants are rich in varieties of secondary metabolites such as flavonoids,
phenolic compounds, alkaloids, and tannins which have been known in vitro to have
antimicrobial properties. Antimicrobial agents are particularly important in decreas-
ing the global effects of infectious diseases [128, 129]. In addition, increasing resis-
tance to traditional antibiotics by pathogenic microbes has made it inevitable to
search for new effective antimicrobial agents of plant origin. Initial examination of
plants for antimicrobial effects begins with the use of crude extracts that are water
1068 M. Z. Kocak and M. G. Kaysim
or alcohol. Antibacterial agents must meet the following conditions to ensure their
action against bacteria: (i) there must be a sensitive antibacterial purpose in the cell,
(ii) antibacterial agents should reach the target in sufficient quantity, and (iii) anti-
bacterial agents should be left to their natural state [130–133].
Anti-inflammatory
Phenolic and flavonoid compounds present in plants have showed important phar-
macological activities, such as anti-inflammatory effects through different mecha-
nisms [134]. Previous studies have shown that inflammation is mostly caused by
damage to living tissues by viral, bacterial, and fungal infections. The fundamental
purpose of inflammatory treatments is to destroy the harmful agents. Furthermore,
nonsteroidal anti-inflammatory drugs such as the isoform of the cyclooxygenase
enzyme (COX-1, COX-2) can reduce pain and inflammation by reducing prosta-
glandin production by inhibiting arachidonic acid metabolism [135]. Although non-
steroidal drugs have many side effects, there are plants (medicinal) that have no side
effects and have significant anti-inflammatory therapeutic effects [136–138].
In the recent years, bibliometric analysis researches have been carried out in various
fields ranging from biology to agriculture and from management-economics to the
communication studies [139–142]. Herewith the bibliometric analysis, the relevant
former studies in certain and specified topics have been discussed and then recom-
mendations as “future outlook” are proposed for the readers. In addition to the con-
ventional bibliometric analysis, in the last decade, a great interest in uses of a
software named “VOSviewer” (developed by Leiden University; for more details,
please visit https://www.vosviewer.com/) has been addressed in innumerable topics
[143–146]. Along with the current chapter, VOSviewer-aided constructed visualiza-
tions were proposed. With the relevant visualizations, such as term analysis (Fig. 1),
keyword analysis (Fig. 2), core-hot keyword analysis (Fig. 3), and country analysis
(Fig. 4), we extracted and deduced some common themes according to the retrieved
data from Scopus, which was of reliable scientific databases [140]. Due to large data
in the relevant field, we used some limitations, viz., medicinal plants (in abstract,
title, keywords on Scopus). As expected, we had a great number of documents,
which were estimated to be about 700,000 but without any refinements. To be com-
patible with the book project, we specified our search using pharmacology, toxicol-
ogy, and pharmaceuticals and then we constructed the relevant figures, in this regard.
Term Analysis of the Documents
Of the analysis conducted, term analysis clearly revealed three major clusters. To
start with the red cluster, it might be “weird” to be “hot-spot” of the word “review.”
Appraisal of Medicinal Plants for Pharmacological Properties 1069
When we have a quick number of document type, we observed that almost all of the
documents were “review papers” (equivalent to 20–20% of 2715 document num-
bers). The green cluster is based on biological activities, assays, and solvents of
extractions. Up to the best knowledge of the original researches, most reports of
herbal medicines are issued on biological activities, in particular antioxidant and
antimicrobial activities.
Keyword Analysis
In relation or comparison to the term analysis, regarding keyword analysis, a high
number of clusters were observed according to our inserted criteria. However, the
commonly and widely used words were “medicinal plants, natural products, anti-
oxidant activities, flavonoids, and cytotoxicity.” Of the metabolites available in
medicinal plants, secondary metabolites (rather than primary metabolites common
to all living organisms) are substantial in well-known activities of the plants. There
are about 100,000 elucidated compounds and flavonoids, a subgroup of secondary
metabolites, as well as hot-keyword of the current study, which are not only and
unique of the metabolites. Even being in a numerous reports, flavonoids are critical
in exerting the activities [147–152], and terpenoids, alkaloids, and other groups of
phenolics are furthermore significant and deserve to be investigated in detail.
Appraisal of Medicinal Plants for Pharmacological Properties 1071
Country Analysis
Corresponding to the country analysis, as expected from all fields, China, India, the
USA, Italy, Brazil, Iran, Germany, Turkey, and Pakistan were noted as the leading
countries in the field medicinal plants according to the number of documents pub-
lished on Scopus. It is significant to note that the number of documents does not
always reflect the quality. The number of documents is, in general, dependent on the
plant species distributed in the relevant region, number of experts in specific topics,
and most significantly laboratory infrastructure. We should also note that a low ratio
of the recorded plant species is assessed for medicinal purposes, being 50,000–80,000
plant species (equivalent to 20% of 400,000 plant species). In this regard, to an
extent, we could not generalize that the number of medicinal plants is directly linked
to the number of plant species. We should also address some issues on the export
and import of the species such as spice, supplementation, food additives, and drugs.
When we should have a quick look on the country-wise and value-wise export of
medicinal herbs (https://pib.gov.in/PressReleasePage.aspx?PRID=1594929), and
the USA, Germany, Vietnam, China, Italy, Pakistan, Bangladesh, the UK, Australia,
and France are the leading countries. As seen from the order of countries, we cannot
1072 M. Z. Kocak and M. G. Kaysim
Generally, medicinal plants and plant products have a promising future, as there are
about half a million of plant species in the world. However, the medical activities of
many of them have not yet been investigated and their medical activities may be
decisive in the treatment of current or future studies. In the same medicinal plant
species, different varieties can provide different amounts of flavonoid and phenolic
compounds as well as biological activities. Therefore, medicinal plant varieties
should be considered for future medical and pharmaceutical research studies.
3 Conclusions
To explore and advance these alternative choices for the use of phytochemical com-
pounds, investigation of medicinal plants along with intensive profiling research is
required. Targeted compounds should be used in biomedical and pharmaceutical
research consisting of in vitro, in vivo, and clinical trial steps to evaluate the safety,
efficacy, as well as side effects of the candidate compounds tested.
References
1. Adhikari PP, Paul SB (2018) History of Indian traditional medicine: a medical inheritance.
Asian J Pharm Clin Res 11:421–426
2. Johns T (2002) Plant genetic diversity and malnutrition: practical steps in the development of
a global strategy linking plant genetic resource conservation and nutrition. Afr J Food Nutr
Sci 3:98–100
3. Khazdair MR, Kianmehr M, Anaeigoudari A (2021) Effects of medicinal plants and flavo-
noids on Parkinson’s disease: a review on basic and clinical evidences. Adv Pharm Bull 11:224
4. Albuquerque UP, do Nascimento ALB, Chaves LS, Feitosa IS, de Moura JMB, Gonçalves P
H S, Júnior WSF. (2020) The chemical ecology approach to modern and early human use of
medicinal plants. Chemoecology 30:89–102
5. Dey A, Mukherjee A, Chaudhury M (2017) Alkaloids from apocynaceae: origin, pharmaco-
therapeutic properties, and structure-activity studies. Stud Nat Prod Chem 52:373–488
6. Hudha MN, Hamidah I, Permanasari A, Abdullah AG, Rachman I, Matsumoto T (2020) Low
Carbon Education: a review and bibliometric analysis. Eur J Educ Res 9:319–329
7. Ahad B, Shahri W, Rasool H, Reshi ZA, Rasool S, Hussain T (2021) Medicinal plants and
herbal drugs: an overview. In: Medicinal and aromatic plants: healthcare and industrial appli-
cations, p 1
8. Singh R (2015) Medicinal plants: a review. J Plant Sci 3:50–55
Appraisal of Medicinal Plants for Pharmacological Properties 1073
29. Gulcin I, Beydemir S (2013) Phenolic compounds as antioxidants: carbonic anhydrase isoen-
zymes inhibitors. Mini Rev Med Chem 13:408–430
30. Astutik S, Pretzsch J, Ndzifon KJ (2019) Asian medicinal plants production and utilization
potentials: a review. Sustainability 11:5483
31. Hosseinzadeh S, Jafarikukhdan A, Hosseini A, Armand R (2015) The application of medicinal
plants in traditional and modern medicine: a review of Thymus vulgaris. Int J Clin Med 6:635
32. Vasisht K, Sharma N, Karan M (2016) Current perspective in the international trade of
medicinal plants materials: an update. Curr Pharm Des 22:4288–4336
33. Bentley R (2010) Medicinal plants. Domville-Fife Press, London, pp 23–46
34. Rao BR, Rajput DK (2010) Global scenario of medicinal plants. World 10500:I800
35. Schıppmann U, Leaman D, Cunnıngham AB (2006) Cultivation and wild collection of
medicinal and aromatic plants under sustainability aspects. In: Bogers RJ, Craker LE, Lange
D (eds) Med aromat plants. Springer, Dordrecht, p 17
36. World Bank (2018) Strategic segmentation analysis: Nepal. Medicinal and aromatic plants.
World Bank Group, Washington, DC, p 61
37. Demir İ (2020) An Ethnobotanical study of medicinal plants used in Hizan district (Bitlis-
Turkey). YYÜ TAR BİL DERG 30:732–741
38. Zahra W, Rai SN, Birla H, Singh S, Sen Rathore AS, Dilnashin H, Keswani C, Singh SP
(2020) Economic importance of medicinal plants in Asian countries. In: Keswani C (ed)
Bioeconomy for sustainable development. Springer, Singapore, pp 359–377
39. Dar RA, Shahnawaz M, Qazi PH (2017) General overview of medicinal plants: a review. J
Phytopharm 6:349–351
40. Jurikova T, Skrovankova S, Mlcek J, Balla S, Snopek L (2018) Bioactive compounds,
antioxidant activity, and biological effects of european cranberry (Vaccinium oxycoccos).
Molecules 24:24
41. Vines G (2004) Herbal harvests with a future: towards sustainable sources for medicinal
plants. Plantlife International
42. Hassan I, Wan Ibrahim WN, Yusuf FM, Ahmad SA, Ahmad S (2020) Biochemical constituent
of ginkgo biloba (Seed) 80% methanol extract inhibits cholinesterase enzymes in javanese
medaka (Oryzias javanicus) model. J Toxicol
43. Kabera J, Semana E, Ally RM, Xin H (2014) Plant secondary metabolites: biosynthesis, clas-
sification, function and pharmacological properties. J Pharm Pharmacol 2:377–392
44. Stamp N (2003) Out of the quagmire of plant defense hypotheses. Q Rev Biol 78:23–55
45. Meng L, Wen KH, Brewin R, Wu Q (2020) Knowledge atlas on the relationship between
urban street space and residents’ health—A bibliometric analysis based on VOSviewer and
vitespace. Sustainability 12:2384
46. Böttger A, Vothknecht U, Bolle C, Wolf A (2018) Plant secondary metabolites and their gen-
eral function in plants. In: Lessons on caffeine, cannabis & co. Springer, Cham, pp 3–17
47. Martínez AL, González-Trujano ME, Aguirre-Hernández E, Moreno J, Soto-Hernández
M, López-Muñoz FJ (2009) Antinociceptive activity of Tilia americana var. mexicana
inflorescences and quercetin in the formalin test and in an arthritic pain model in rats.
Neuropharmacology 56:564–571
48. Alamgir ANM (2017) Cultivation of herbal drugs, biotechnology, and in vitro production
of secondary metabolites, high-value medicinal plants, herbal wealth, and herbal trade. In:
Therapeutic use of medicinal plants and their extracts, vol 1, pp 379–452
49. Hussein RA, El-Anssary AA (2019) Plants secondary metabolites: the key drivers of the
pharmacological actions of medicinal plants. Herb Med 1:13
50. Spiller F, Alves MK, Vieira SM, Carvalho TA, Leite CE, Lunardelli A, Poloni JA, Cunha FQ,
de Oliveira JR (2008) Anti-inflammatory effects of red pepper (Capsicum baccatum) on car-
rageenan and antigen-induced inflammation. J Pharm Pharmacol 60(4):473–478
51. Shilova IV, Krasnov EA, Korotkova EI et al (2006) Antioxidant properties of extracts from
the above-ground parts of Filipendula ulmaria. Pharm Chem J 40:660–662
Appraisal of Medicinal Plants for Pharmacological Properties 1075
73. Alamgir ANM (2018) Secondary metabolites: secondary metabolic products consisting of
C and H; C, H, and O; N, S, and P elements; and O/N heterocycles. In: Therapeutic use of
medicinal plants and their extracts, vol 2, pp 165–309
74. Fakhri S, Moradi SZ, Ash-Rafzadeh A, Bishayee A (2021) Targeting cellular senescence in
cancer by plant secondary metabolites: a systematic review. Pharmacol Res:105961
75. Fernandez-Panchon MS, Villano D, Troncoso AM, Garcia-Parrilla MC (2008) Antioxidant
activity of phenolic compounds: from in vitro results to in vivo evidence. Crit Rev Food Sci
Nutr 48:649–671
76. Marrelli M (2021) Medicinal plants. Plants 10:1355
77. Seca AM, Pinto DC (2018) Plant secondary metabolites as anticancer agents: successes in
clinical trials and therapeutic application. Int J Mol Sci 19:263
78. Erhirhie EO, Emeghebo CN, Ilodigwe EE, Ajaghaku DL, Umeokoli BO, Eze PM, Ngwoke
KG, Chiedu OF (2019) Dryopteris filix-mas (L.) Schott ethanolic leaf extract and fractions
exhibited profound anti-inflammatory activity. Avicenna J Phytomedicine 9:396–409
79. Kumar M, Prakash S, Kumari N, Pundir A, Punia S, Saurabh V, Mekhemar M (2021)
Beneficial role of antioxidant secondary metabolites from medicinal plants in maintaining
oral health. Antioxidants 10:1061
80. Zaynab M, Fatima M, Abbas S, Sharif Y, Umair M, Zafar MH, Bahadar K (2018) Role of
secondary metabolites in plant defense against pathogens. Microb Pathog 124:198–202
81. Manandhar S, Luitel S, Dahal RK (2019) In vitro antimicrobial activity of some medicinal
plants against human pathogenic bacteria. J Trop Med
82. Scharf DH, Heinekamp T, Brakhage AA (2014) Human and plant fungal pathogens: the role
of secondary metabolites. PLoS Pathog 10:e1003859
83. Seca AM, Pinto DC (2019) Biological potential and medical use of secondary metabolites.
Medicines 6:66
84. Kyaw YMM, Bi Y, Oo TN, Yang X (2021) Traditional medicinal plants used by the mon
people in Myanmar. J Ethnopharmacol 265:113253
85. Wink M (2012) Medicinal plants: a source of anti-parasitic secondary metabolites. Molecules
17:12771–12791
86. Alzobaidi N, Quasimi H, Emad NA, Alhalmi A, Naqvi M (2021) Bioactive compounds and
traditional herbal medicine: promising approaches for the treatment of dementia. Degener
Neurol Neuromuscul Dis 11:1
87. Bhat SG (2021) Medicinal plants and its pharmacological values. In: Pharmacognosy-
medicinal plants
88. Pagare S, Bhatia M, Tripathi N, Pagare S, Bansal YK (2015) Secondary metabolites of plants
and their role: overview. Curr Trends Biotechnol Pharm 9:293–304
89. Yang L, Yang C, Li C, Zhao Q, Liu L, Fang X, Chen XY (2016) Recent advances in biosyn-
thesis of bioactive compounds in traditional Chinese medicinal plants. Sci Bull 6:3–17
90. Bartwal A, Mall R, Lohani P, Guru SK, Arora S (2013) Role of secondary metabolites
and brassinosteroids in plant defense against environmental stresses. Plant Growth Regul
32:216–232
91. Irfan A, Imran M, Khalid M, Ullah MS, Khalid N, Assiri MA, Shahzad M (2021) Phenolic
and flavonoid contents in Malva sylvestris and exploration of active drugs as antioxidant
and anti-COVID19 by quantum chemical and molecular docking studies. J Saudi Chem Soc
25:101277
92. Patra JK, Kim ES, Oh K et al (2014) Antibacterial effect of crude extract and metabolites of
Phytolacca americana on pathogens responsible for periodontal inflammatory diseases and
dental caries. BMC Complement Altern Med 14:343
93. Tetali SD (2019) Terpenes and isoprenoids: a wealth of compounds for global use.
Planta 249:1–8
94. Bouyahya A, Belmehdi O, Benjouad A, El Hassani RA, Amzazi S, Dakka N, Bakri Y (2020)
Pharmacological properties and mechanism insights of moroccan anticancer medicinal
plants: what are the next steps? Ind Crops Prod 147:112198
Appraisal of Medicinal Plants for Pharmacological Properties 1077
95. Ng ZX, Koick YTT, Yong PH (2021) Comparative analyses on radical scavenging and cyto-
toxic activity of phenolic and flavonoid content from selected medicinal plants. Nat Prod Res
35:5271–5276
96. Theis N, Lerdau M (2003) The evolution of function in plant secondary metabolites. Int J
Plant Sci 164:S93–S102
97. Abdallah II, Quax WJ (2017) A glimpse into the biosynthesis of terpenoids. KnE Life
Sci:81–98
98. Trost BM, Min C (2020) Total synthesis of terpenes via palladium-catalysed cyclization strat-
egy. Nat Chem 12:568–573
99. Malekmohammad K, Rafieian-Kopaei M (2021) Mechanistic aspects of medicinal plants
and secondary metabolites against severe acute respiratory syndrome coronavirus 2 (SARS-
CoV-2). Curr Pharm Des
100. Dey P, Kundu A, Kumar A, Gupta M, Lee BM, Bhakta T, Kim HS (2020) Analysis of alka-
loids (indole alkaloids, isoquinoline alkaloids, tropane alkaloids). In: Recent advances in
natural products analysis, pp 505–567
101. Duan SC, Kwon SJ, Eom SH (2021) Effect of thermal processing on color, pheno-
lic compounds, and antioxidant activity of Faba bean (Vicia faba L.) leaves and seeds.
Antioxidants 10:1207
102. Munir N, Iqbal AS, Altaf I, Bashir R, Sharif N, Saleem F, Naz S (2014) Evaluation of anti-
oxidant and antimicrobial potential of two endangered plant species Atropa belladonna and
Matricaria chamomilla. Afr J Trad Comp Alter Med: AJTCAM 11:111–117
103. Šafratová M, Hošťálková A, Hulcová D, Breiterová K, Hrabcová V, Machado M, Cahlíková
L (2018) Alkaloids from Narcissus poeticus cv. Pink parasol of various structural types and
their biological activity. Arch Pharm Res 41:208–218
104. Jamshidi-Kia F, Lorigooini Z, Amini-Khoei H (2018) Medicinal plants: past history and
future perspective. J HerbMed Pharmacol:7
105. Twilley D, Lall N (2018) The role of natural products from plants in the development of
anticancer agents. Nat Prod Res:139–178
106. Christl H, Morilla J, Hoen T, Zumkier U (2019) Comparative assessment of the intrinsic
sensitivity of crop species and wild plant species to plant protection products and their active
substances and potential implications for the risk assessment: a literature review. Integr
Environ Assess Manag 15:176–189
107. Dajic-Stevanovic Z, Pljevljakusic D (2015) Challenges and decision making in cultivation of
medicinal and aromatic plants. Int J Med Aromat Plants. Springer, Dordrecht:145–164
108. Ulewicz-Magulska B, Wesolowski M (2019) Total phenolic contents and antioxidant poten-
tial of herbs used for medical and culinary purposes. Plant Foods Hum Nutr 74:61–67
109. Tungmunnithum D, Thongboonyou A, Pholboon A, Yangsabai A (2018) Flavonoids and
other phenolic compounds from medicinal plants for pharmaceutical and medical aspects:
an overview. Medicines 5:93
110. Ansari MA, Anurag A, Fatima Z, Hameed S (2013) Natural phenolic compounds: a potential
antifungal agent. Microbial pathogens and strategies for combating them. Sci Technol Educ
1:1189–1195
111. Keswani C, Mishra S, Sarma BK, Singh SP, Singh HB (2014) Unraveling the efficient appli-
cations of secondary metabolites of various Trichoderma spp. Appl Microbiol Biotechnol
98:533–544
112. Roleira FM, Varela CL, Costa SC, Tavares-da-Silva EJ (2018) Phenolic derivatives from
medicinal herbs and plant extracts: anticancer effects and synthetic approaches to modulate
biological activity. Stud Nat Prod Chem 57:115–156
113. Taslimi P, Gulçin İ (2017) Antidiabetic potential: in vitro inhibition effects of some natu-
ral phenolic compounds on α-glycosidase and α-amylase enzymes. J Biochem Mol Toxicol
31:e21956
114. Gurib-Fakim A (2006) Medicinal plants: traditions of yesterday and drugs of tomorrow. Mol
Aspects Med 27:1–93
1078 M. Z. Kocak and M. G. Kaysim
115. Pham-Huy LA, He H, Pham-Huy C (2008) Free radicals, antioxidants in disease and health.
Int J Biomed Sci: IJBS 4:89
116. Adebayo SA, Dzoyem JP, Shai LJ, Eloff JN (2015) The anti-inflammatory and antioxidant
activity of 25 plant species used traditionally to treat pain in southern African. BMC Complt
Altern Med 15:1–10
117. Akinyemi O, Oyewole SO, Jimoh KA (2018) Medicinal plants and sustainable human health:
a review. Hortic Int J 2:194–195
118. Škrovánková S, Mišurcová L, Machů L (2012) Antioxidant activity and protecting health
effects of common medicinal plants. Adv Food Nutr Res 67:75–139
119. Karakaya S, Koca M, Sytar O, Duman H (2020) The natural phenolic compounds and their
antioxidant and anticholinesterase potential of herb Leiotulus dasyanthus (K. Koch) Pimenov
& Ostr. Nat Prod Res 34:1303–1305
120. Boulmokh Y, Belguidoum K, Meddour F, Amira-Guebailia H (2021) Investigation of anti-
oxidant activity of epigallocatechin gallate and epicatechin as compared to resveratrol and
ascorbic acid: experimental and theoretical insights. Struct Chem:1–17
121. Ninkuu V, Zhang L, Yan J, Fu Z, Yang T, Zeng H (2021) Biochemistry of terpenes and recent
advances in plant protection. Int J Mol Sci 22:5710
122. Kabera JN, Semana E, Mussa AR, He X (2014) Plant secondary metabolites: biosynthesis,
classification, function and pharmacological properties. J Pharm Pharmacol 2:377–392
123. Nadon B, Jackson S (2020) The polyploid origins of crop genomes and their implications: a
case study in legumes. Adv Agron 159:275–313
124. Duffner A, Moser T, Candolfi MP (2020) Feasibility of assessing vegetative and genera-
tive endpoints of crop-and non-crop terrestrial plant species for non-target terrestrial plant
(NTTP) regulatory testing under greenhouse conditions. PloS One 15:e0230155
125. Frimpong EK, Asong JA, Aremu AO (2021) A review on medicinal plants used in the man-
agement of headache in Africa. Plants 10:2038
126. Tavallali H, Bahmanzadegan A, Rowshan V, Tavallali V (2021) Essential oil composition,
antioxidant activity, phenolic compounds, Total phenolic and flavonoid contents from pom-
ace of Citrus aurantifolia. JMPB 10:103–116
127. Traore KF, Kone KY, Ahi AP, Soro D, Assidjo NE, Fauconnier ML, Sindic M (2021) Phenolic
compounds characterisation and antioxidant activity of black plum (Vitex doniana) fruit pulp
and peel from Côte d’Ivoire. J Food Meas Charact 15:1281–1293
128. Manoharachary C, Nagaraju D (2016) Medicinal plants for human health and welfare. Ann
Phytomedicine 5:24–34
129. Mou Z, Zhao Y, Ye F, Shi Y, Kennelly EJ, Chen S, Zhao D (2021) Identification, biological
activities and biosynthetic pathway of dendrobium alkaloids. Front Pharmacol:12
130. Oualcadi Y, Aityoub A, Berrekhis F (2021) Investigation of different antioxidant capacity
measurements suitable for bioactive compounds applied to medicinal plants. J Food Meas
Charact 15:71–83
131. Romulo A, Zuhud EA, Rondevaldova J, Kokoska L (2018) Screening of in vitro antimicrobial
activity of plants used in traditional Indonesian medicine. Pharm Biol 56:287–293
132. Vatľák A, Kolesárová A, Vukovič N, Rovná K, Petrová J, Vimmerová V, Kačániová M (2021)
Antimicrobial activity of medicinal plants against different strains of bacteria. J Microbiol
Biotechnol Food Sci:174–176
133. Whangchai K, Shanmugam S, Van Le Q, Chau TP, Al-Kheraif AA, Brindhadevi K, Duc PA
(2021) Study of antimicrobial activity of Thespesia populnea-coated nanozirconium on cot-
ton gauze fabrics. Appl Nanosci:1–7
134. Puangpraphant S, Cuevas-Rodríguez EO, Oseguera-Toledo M (2022) Anti-inflammatory and
antioxidant phenolic compounds. In: Current advances for development of functional foods
modulating inflammation and oxidative stress. Academic Press, pp 165–180
135. Schultz F, Osuji OF, Wack B, Anywar G, Garbe LA (2021) Antiinflammatory medicinal
plants from the Ugandan greater mpigi region act as potent inhibitors in the COX-2/PGH2
pathway. Plants 10:351
Appraisal of Medicinal Plants for Pharmacological Properties 1079
136. Kuete V (2014) Health effects of alkaloids from african medicinal plants. In: Kuete V (ed)
Toxicological survey of African medicinal plants, pp 611–633
137. Osungunna MO (2021) Screening of medicinal plants for antimicrobial activity: pharmacog-
nosy and microbiological perspectives. J Microbiol Biotechnol:727–735
138. Ribeiro VP, Arruda C, Abd El-Salam M, Bastos JK (2018) Brazilian medicinal plants with
corroborated anti-inflammatory activities: a review. Pharm Biol 56:253–268
139. Büyükbaykal NG, İli B (2021) E-Spor kavramına yönelik araştırmaların bibliyometrik anal-
izi bibliometric analysis of researches for e-sport. UKSAD 6:572–583
140. Celik E, Durmus A, Adizel O, Uyar HN (2021) A bibliometric analysis: what do we know
about metals (loids) accumulation in wild birds? Environ Sci Pollut Res 28:10302–10334
141. Ritter MR, Silva TCD, Araújo EDL, Albuquerque UP (2015) Bibliometric analysis of ethno-
botanical research in Brazil (1988–2013). Acta Bot Brasilica 29:113–119
142. Yeung AWK, Heinrich M, Atanasov AG (2018) Ethnopharmacologya bibliometric analysis
of a field of research meandering between medicine and food science? Front Pharmacol 9:215
143. Hussein RA, El-Anssary AA (2019) Plants secondary metabolites: the key drivers of the
pharmacological actions of medicinal plants. J Herb Med 1:13
144. Kumar S (2017) Plant secondary metabolites (PSMs) of Brassicaceae and their role in plant
defense against insect herbivores – a review. J Appl Nat Sci 9:508–519
145. Mickymaray S (2019) Efficacy and mechanism of traditional medicinal plants and bioactive
compounds against clinically important pathogens. Antibiotics 8:257
146. Yu Y, Li Y, Zhang Z, Gu Z, Zhong H, Zha Q, Chen E (2020) A bibliometric analysis using
VOSviewer of publications on COVID-19. Ann Transl Med:8
147. Isah T (2019) Stress and defense responses in plant secondary metabolites production.
Biol Res 52
148. Khumalo GP, Van Wyk BE, Feng Y, Cock IE (2022) A review of the traditional use of
Southern African medicinal plants for the treatment of inflammation and inflammatory pain.
J Ethnopharmacol 283:114436
149. Quílez M, Ferreres F, López-Miranda S, Salazar E, Jordán MJ (2020) Seed oil from
Mediterranean aromatic and medicinal plants of the Lamiaceae family as a source of bioac-
tive components with nutritional. Antioxidants 9:510
150. Savych A, Milian I (2021) Total flavonoid content in the herbal mixture with antidiabetic
activity. PharmacologyonLine 2:68–75
151. Solnier J, Fladerer JP (2021) Flavonoids: a complementary approach to conventional therapy
of COVID-19? Phytochem Rev 20:773–795
152. Venkataramaiah C, Payani S, Priya BL, Pradeepkiran JA (2021) Therapeutic potentiality
of a new flavonoid against ketamine induced glutamatergic dysregulation in schizophrenia:
in vivo and in silico approach. Biomed Pharmacother 138:111453
Pharmacological Properties and Tissue
Culture Method of Endangered Medicinal
Plants
1 Introduction
China is one of the richest countries in the world in terms of plant species, with
more than 30,000 species of higher plants alone, accounting for about 10% of the
world’s species, including more than 11,000 species of medicinal plants. The 2020
edition of the Chinese Pharmacopoeia includes 602 species of medicinal origin
plants [1]. Only about half of these medicinal plant species have been propagated
and cultivated artificially, while the other half rely on the harvesting and processing
of wild resources for medicine. Due to long-term overharvesting and economic and
social development breaking the balance of the ecosystem, the wild resources of
some medicinal plants are becoming increasingly depleted. The Red Book of
Chinese Plants-Rare and Endangered Plants contains 168 species of medicinal
plants, accounting for 43.30% of all species, including 46 species of commonly
used traditional Chinese medicines and 19 species of bulk medicinal herbs (a total
of 27 origin plants) [2]. These rare and endangered medicinal plants share common
botanical characteristics, such as narrow distribution, slow growth rate, low fruit set,
Y. Zheng
Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of
Botany, Jiangsu Province and Chinese Academy of Sciences (Nanjing Botanical Garden
Mem. Sun Yat-Sen), Nanjing, People’s Republic of China
X. Shi
Ningxia Institute of Quality Standards and Testing Technology for Agricultural Products,
Yinchuan, People’s Republic of China
L. Fu (*)
Key Laboratory of Novel Materials for Sensor of Zhejiang Province, College of Materials and
Environmental Engineering, Hangzhou Dianzi University,
Hangzhou, People’s Republic of China
e-mail: [email protected]
poor seed development, or difficulty in seed germination. These reasons cause their
natural populations to renew slowly and become rare and endangered. Therefore,
the development of artificial propagation techniques and population conservation
are prerequisites for the sustainable exploitation of such plants. This chapter will
provide an overview of the current status of research and conservation of propaga-
tion techniques for rare and endangered medicinal plants.
Among the methods of artificial propagation, seed propagation is undoubtedly the
easiest method. However, most of the rare and endangered medicinal plants have spe-
cial breeding systems and seed structures. They usually have long germination cycles
and low germination rates. Therefore, studying the biological properties and germina-
tion characteristics of seeds to improve the germination rate is the most important
research content and direction of seed propagation. Grafting propagation is only appli-
cable to woody rare and endangered medicinal plants, although the speed of seedling,
but the reproduction factor is not high. Its survival rate is greatly influenced by the age
of the scion and its affinity with the rootstock. Propagation by cuttings is suitable for all
woody and some herbaceous rare and endangered medicinal plants, with a larger repro-
duction factor but a longer seedling generation period. Its survival rate is influenced by
the season of cuttings, cutting substrate, age of the spike, and the type and concentra-
tion of rooting agent. Rapid propagation by tissue culture is a method applicable to all
cherished and endangered medicinal plants for rapid propagation. It has a high repro-
duction coefficient and fast reproduction rate and is especially suitable for rapid repro-
duction on a large scale and factory scale. It is the most intensively and extensively
studied method of rapid propagation of rare and endangered medicinal plants. However,
it is not easy to establish an efficient and stable technical system because the character-
istics of different species of plants themselves vary greatly.
2 Seed Propagation
Inefficient seed reproduction is one of the most direct causes of plant rarity and
endangerment. The main factors that make seed reproduction difficult are the fol-
lowing: (1) low fruit set due to special pollination methods; (2) long post-maturation
period of seeds; (3) presence of seed germination inhibitors; and (4) difficulty in
germination due to special seed structure. In order to improve the ability of rare and
endangered plants to reproduce sexually, research has been conducted on their own
botanical characteristics.
In the wild, orchids require specific pollinating insects to assist in pollination. Due
to the low number of pollinators or the special flower structure that cannot attract
pollinators, the Orchidaceae has only about 5% fruit set rate [3]. Combined with the
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1083
fact that the seed embryo of is Orchidaceae a multicellular primitive embryo body
and has no endosperm, most orchids have low seed germination rates [4–6]. Yagame
et al. [7] isolated symbiotic bacteria from Cremastra appendiculata that promote
seed germination, thus speculating that symbiotic germination may be the best way
to germinate seeds of Cremastra appendiculata. Unlike the orchids, the seeds of the
woody plant Magnolia officinalis varied in seed weight between 0.1 and 0.28 g in
different seed zones and in different orientation canopies [8–10]. Seed size was
found to be positively correlated with germination ability after grading seeds
according to single seed weight. The germination rate and average daily germina-
tion rate varied very much between grades. Differences in the contents of soluble
sugars, soluble proteins, and crude fats of the seeds of different grades during stor-
age also reached significant levels. Growth indicators such as mortality, seedling
height, and ground diameter of seedlings from large seeds were also significantly
better than those of small seeds.
The Tertiary relict Cercidiphyllum japonicum is a dioecious plant with a frag-
mented population distribution resulting in a low fruiting rate [11]. At the same
time, its seeds are small (only about 2.0 mm in width and 1.5 mm in length), which
also leads to low seed reproduction rate. Dosmann et al. [12, 13] chilling seeds at
3.5 °C for 8 d and placing them on a paper sprout bed at 25 °C in darkness or 15 h
of light increased their germination rate from 42% to 75%. A certain concentration
of gibberellin solution can also improve the germination rate of Cercidiphyllum
japonicum [14]. Ephedra sinica is a species in the Ephedraceae, with a long seed
development cycle, taking about 3 months from flowering to seed maturity. Although
it has a large number of flowers, but the flower defeat rate is high, and the flowers
and fruits are all easy to fall off, resulting in its low seed yield and poor quality [15,
16]. In addition, the thick and dense seed coat of Ephedra sinica hinders water
uptake during germination and reduces the rate of gas exchange. The presence of
germination-inhibiting substances within the seeds also contributes to the germina-
tion rate. Both the use of low-temperature lamination and treatment with gibberellin
significantly improved seed germination, resulting in 94% and 90% germination,
respectively, nearly three times higher than the control [17].
water coupled with refrigeration at 5 °C for 14 d was the most effective method to
break its dormancy, but the germination rate was only 26.1%. The seeds of the
genus Taxus have the phenomenon of after-ripening [26]. Germination inhibitors
are unevenly present in the lignified seed coat and are poorly permeable to water
and air [27, 28]. Therefore, the germination cycle is long in the natural state, usually
taking at least one year to germinate [29]. Panax ginseng is mainly propagated by
seeds, but the seeds have post-maturation characteristics and require long morpho-
logical and physiological maturation before germination. Some studies have shown
that gibberellin can promote Panax ginseng seed cleavage but not embryo growth
[30–32].
Among the factors contributing to the long germination cycle of seeds of rare and
endangered medicinal plants, the widespread presence of germination inhibitors is
one of the most important factors. Such substances may be organic acids, alkaloids,
or some organic substances that release ammonia or cyanide upon decomposition.
They may be present in any part of the seed or fruit, and only when these inhibitory
substances are eliminated, can the seed germinate properly.
Rhodiola rosea substantially increased its germination rate either in 0–5 °C envi-
ronment or treated with 50–1000 mg/L gibberellin [33]. Low temperature and GA
relieved dormancy, thus promoting seed germination. The methanolic extract of
Rauvolfia yunnanensis seed kernels significantly reduced the germination rate of
Brassica campestris seeds, indicating the presence of germination inhibitors in the
seeds [34]. The germination rate of Rauvolfia yunnanensis seeds was significantly
increased up to more than 80% when the seeds were immersed at 28 °C for 24 h
under 200–2000 mg/L gibberellin. Both Cistanche deserticola and Cynomorium
songaricum are parasitic plants, and both have similar seed structures. The seed
embryo is a spherical protoembryo without differentiation of germ, radicle, and
cotyledons. The seeds contain the germination-inhibiting substance abscisic acid
[35–37]. Both seeds can be subjected to sufficient rainfall and can be showered with
germination-inhibiting material or finish post-maturing by lamination [37, 38]. Like
Cynomorium songaricum, Cistanche deserticola seed germination requires the pro-
duction of “shoot tubular organs” or “radicle-like structures” under the action of a
germination-stimulating substance released from the roots of the host plant. The end
of this structure expands to form primary suckers, which adhere to the surface of the
host root, then invade the host root, and connect with the vascular bundle of the host
plant to produce secondary suckers to complete the parasitic process [39–41]. In
their natural state, the chances of encountering host roots after the seeds of
Cynomorium songaricum and Cistanche deserticola mature are very small, which is
an important reason why these wild herbs are on the verge of depletion.
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1085
3 Graft Propagation
The transfer of a part of a plant nutrient organ (branch, bud, scion, scion bud) to
another plant (rootstock) is to use interspecies affinity to make the scion and root-
stock heal at the formation layer of the union, so that the ducts and sieve tubes
interoperate and develop into a new individual. Therefore, the selection of scion and
rootstock is a key factor in the success of grafting. Compared with other asexual
propagation methods, grafting propagation has a low reproduction coefficient, but
the cycle of seedling establishment is short. Grafting is one of the most effective
methods of asexual propagation for woody plants that have difficulty cuttings
rooting.
Peng and Wang [46] developed a prediction model for the survival rate of
Eucommia ulmoides grafting using a quantitative regression method. The results
showed that the age and budding status of the mother plant were important factors
affecting the survival rate of Eucommia ulmoides grafting. Fan [47] found that the
thickness of the rootstock also had a significant effect on the growth of grafted
sprouting branches after grafting. Zhou et al. [48] found that with live seedlings of
Magnolia officinalis as rootstock, the survival rate of Magnolia officinalis popula-
tions from different sources differed significantly after grafting, as well as the
growth of branches and leaves after survival. In the grafting propagation of
Camptotheca acuminata, different sizes of scions had highly significant effects on
the survival rate of grafting and the growth of seedling height and diameter at breast
height of grafted seedlings [49]. For grafting of Torreya grandis, annual branches of
15 cm in length and 0.4 cm in diameter were selected, and the diameter of the scion
and rootstock were nearly equal [50]. The most suitable time for grafting is from
mid-February to early March to ensure a high survival rate. A study on grafting
seedlings of Taxus chinensis with 6-year-old rootstock and 1-year-old semi-woody
branches as scions found that when the scion was grafted in mid-February, the heal-
ing rate and tapping rate of the scion were highest. The tapping length was the
1086 Y. Zheng et al.
longest as well. The growth of the grafted seedlings was best when the scion was
grafted in mid-February and the buds were full and close to the interface. If the
3-year-old seedlings as rootstock, the scion with 1 leaf of the main tip of the reces-
sive axillary bud for the scion can promote the scion into bud early, improve the
survival rate of the main tip, and can form a complete crown shape. If you use
3-year-old seedlings and with a leaf of the main tip of the recessive axillary buds for
rootstock and scion, respectively, can promote the scion early into bud, improve the
survival rate of the main tip, and can form a complete crown shape [51].
4 Cutting Propagation
Propagation from cuttings is one of the most common ways of asexual propaga-
tion. The process involves intercepting a part of a plant’s nutritional organ, spray-
ing or dipping it in a certain concentration of rooting agent, and inserting it into a
loose and moist cultivation medium to make it root and branch and develop into a
complete plant. According to the source of the spike can also be divided into
branch cuttings, root cuttings, bud cuttings, and leaf cuttings. Compared with
other asexual propagation methods, the reproduction coefficient of cuttings is
high, fast, and easy to operate. The cuttings are generally semi-woody branches,
and the cuttings require a loose, water-holding medium. The season of cuttings
and the choice of rooting agent concentration are the most influential factors in
the survival rate of cuttings due to the great variation in the biological character-
istics of the plants themselves.
Chen et al. [52] showed that the most suitable period for cutting rooting of
Eucommia ulmoides cuttings in Beijing is from late May to mid-July. The survival
rate of 2-year-old young tree shoot for cutting can reach 60%, while the survival
rate of adult tree shoot for cutting is less than 20%. The rooting agents were effec-
tive in inducing adventitious root growth of the shoot. Among them, naphthale-
neacetic acid 50 μg/mL had the highest rooting rate. Both NAA and indolebutyric
acid promoted the rooting of Cercidiphyllum japonic shoot cuttings by combining
various indices such as rooting time, average root number, and root length [53].
IBA 100 mg/L + NAA 100 mg/L soaking the cuttings for 4 h was the most suitable
way to take cuttings. Since 2004, Fu et al. [54–59] conducted a systematic study
on the fast propagation technology of Taxus wallichiana var. mairei cuttings,
which involved the composition of cutting substrates, selection of spikes, types
and concentrations of rooting agents, the influence of factors such as cutting sea-
son on rooting, the management of light, fertilizer, pest control, and the construc-
tion of harvesting beds for cuttings. These works have laid the technical foundation
for the establishment of the rapid propagation technology system of cuttings of
Taxus wallichiana var. mairei. On this basis, Taxus cuspidata and Taxus × media
from the same genus have also achieved rapid reproduction of cuttings on a
large scale.
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1087
Here, tissue culture is defined as the inoculation of a part of the plant body (i.e., the
explant, which can be a part or a whole organ), by aseptic manipulation, onto a
culture medium and cultivation in artificially controlled conditions to produce a
complete plant. Compared with other fast propagation methods, tissue culture
requires fewer plants, has a high reproduction coefficient, and is fast, which is the
main way to achieve large-scale fast propagation of rare and endangered plants.
According to whether to form healing tissue, group culture fast breeding can be
divided into two types: one is to induce exosomes to produce healing tissue and
proliferate, forming somatic embryos, and then differentiate from somatic embryos
to grow small plants (or direct differentiation of clumped shoots from healing tissue
after clumped shoot induction culture). This method of reproduction has the highest
reproduction coefficient and the fastest rate. The other type induces buds directly
from the explants without healing tissue induction and undergoes clumped bud
induction culture. Regardless of the route, the selection and sterilization of the
explants and the type and concentration of growth regulators used are crucial to the
success of fast breeding.
Chakraborty et al. [60] used Podophyllum hexandrum roots and rhizomes as
explants and Murashige-Skoog medium as the basic medium to induce clumped
shoots from the roots and rhizomes of Podophyllum hexandrum by adding cytokinin
KT and growth hormone 2,4-D, and eventually obtained sterile seedlings. Rajesh
et al. [61] inoculated the seed embryos of Podophyllum hexandrum onto MS medium
supplemented with 2,4-D (1.5 mg/L) and obtained embryonic healing tissue, which
was regulated by the addition of ABA to obtain a large number of somatic embryos.
After further differentiation and culture, the somatic embryos regenerated plantlets,
establishing a technical system for the regeneration of Peachy ginseng with seed
embryo explants. Tetrastigma hemsleyanum fast propagation can be carried out
using axillary buds, stem tips, and also young sprouted stem segments as explants
[62–64]. The low light is favorable for the sprouting of axillary buds [65–75]. The
reproduction factor of Dendrobium fimbriatum can be 5–7 times higher by using the
lateral buds and stem segments as explants, respectively, for group culture fast prop-
agation [76, 77]. The addition of certain concentrations of polyconazole and lantha-
num nitrate shortened the time of regenerative shoot differentiation of Dendrobium
fimbriatum test tube seedlings and increased the proliferation rate of histopatho-
genic seedlings. This treatment also promoted root differentiation, increased the
number of roots, and improved the rooting rate [78, 79].
The fast propagation of basal plant Anoectochilus roxburghii, Anoectochilus for-
mosanus, and Anoectochilus chapaensis of another group of medicinal plants of the
Orchidaceae has also been systematically studied [80–87]. They can be fastidiously
propagated by inducing clumped shoots from seeds after sterile germination. They
can also be used as explants with sprouted stem segments, stem tips, or stem pieces
about 2 mm thick to induce the production of adventitious shoots and thus obtain
clumped shoots for fast propagation.
1088 Y. Zheng et al.
In situ conservation refers to the protection of valuable ecosystems and wildlife and
their habitats by means of various types of nature reserves and national parks. This
approach is used to maintain the reproduction and evolution of organisms and the
flow of material and energy and ecological processes within the ecosystem. The
principle of in situ conservation is to achieve the conservation of endangered spe-
cies through the conservation of the ecological environment and the integrity of
biological taxa [88, 89]. It is the preferred measure for the conservation of rare and
endangered species. National parks are another form of protected area that first
originated in the United States and has since been adopted by most countries and
regions around the world [90]. The establishment of national parks is an important
element in the construction of Chinese ecological civilization system, which can
protect the originality and integrity of natural ecology and natural cultural heritage
[90, 91]. It provides stricter protection for important ecosystems, while providing
long-term protection for rare wildlife and plants [92–94]. As of 2020, China has
established 10 national park, including Sanjiangyuan, Hainan Tropical Rainforest,
and Shennongjia, with a total area of 222,900 square kilometers. The established
nature reserves and national parks have enabled the effective protection of almost all
rare and endangered species on the Red Book of Rare and Endangered Plants of
China, including rare and endangered medicinal plants.
Ex situ conservation refers to the relocation of species whose survival and reproduc-
tion are seriously threatened due to the non-existence of living conditions and very
low numbers of species out of their original locations to zoos, botanical gardens, or
breeding centers for endangered plants and animals for conservation and manage-
ment [95, 96]. Ex situ conservation is a useful complement to in situ conservation
and is an important part of biodiversity conservation [97]. Ex situ conservation can
temporarily preserve and protect a certain number of populations for a short period
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1089
of time [98]. For rare and endangered medicinal plants, botanical gardens, arbore-
tums, or medicinal plant gardens are the ideal places for their ex situ conservation
[99, 100]. The construction level of Chinese botanical gardens and medicinal botan-
ical gardens is close to the level of developed countries. At present, there are 162
botanical gardens and arboretums in China, with a total area of 102,007 hectares.
These botanical and arboretum gardens protect 42.5% of the rare and endangered
plants in the Red Book of Rare and Endangered Plants of China.
At present, there is a contradiction in the development of Chinese herbal medi-
cine industry: On the one hand, there is a serious lack of raw materials for products
based on rare and endangered plants in the herbal medicine market [101–103]. On
the other hand, a lot of human and material resources are needed for the ex situ and
in situ conservation of rare and endangered medicinal plants. From the above review,
it is clear that the most important reason for the endangerment of rare and endan-
gered plants is that their own breeding systems are somehow defective [104, 105].
If we can strengthen the research of artificial propagation and supporting cultivation
technology of rare and endangered medicinal plants while protecting them in situ
and ex situ them, we can not only provide sufficient raw materials for herbs and
medicines, but also make rare and endangered medicinal plants protected or even
get out of the endangered status.
7 Conclusion
Beginning with the establishment of nature reserves in the 1950s to protect rare and
endangered medicinal plants in situ, research on rare and endangered medicinal
plants has been conducted in China for more than 70 years. Botanists have studied
the reproductive biology of many rare and endangered plants and have also estab-
lished a system of artificial breeding techniques for many rare and endangered
plants. The systematic study of the reproductive biology of orchids found that the
pollination of orchids is dominated by insect vectors, but some taxa lack sufficient
pollinators due to the low number of pollinating insects in their distribution sites or
the special flower structure and thus have low fruit set rates. In addition, the seed
embryos of some orchids are incompletely developed and have no endosperm, so
there is a long germination cycle due to the phenomenon of post-maturation. At the
same time, the supply of nutrients for seed germination is not guaranteed, resulting
in low germination rate. Besides, symbiotic fungi were also isolated from sme rare
and endangered orchid medicinal plants, and it is speculated that they may have
symbiotic germinahion. Based on the results of these |studies, targeted strategies
were developed to greatly improve the germination rate and reproduction ability of
rare and endangered medicinal plants of the orchid family and laid the foundation
for the development and utilization of rare and endangered medicinal plants of the
Orchidaceae. It should be noted, however, that there are many rare and endangered
plants whose reproductive biology and propagation methods have not been studied
systematically and thoroughly enough. Therefore, they have not been established a
1090 Y. Zheng et al.
proven artificial breeding system, such as the rare and endangered medicinal fern,
Cibotium barometz, is one of them. Therefore, we still have much work to do to
strengthen the research, protection, development, and utilization of rare medicinal
plant and animal resources and to promote the development of China’s traditional
Chinese medicine industry.
References
18. Tasheva K, Kosturkova G (2012) The role of biotechnology for conservation and biologically
active substances production of Rhodiola rosea: endangered medicinal species. Sci World J
2012:274942. https://doi.org/10.1100/2012/274942
19. Karimi-Maleh H, Orooji Y, Karimi F, Alizadeh M, Baghayeri M, Rouhi J, Tajik S, Beitollahi
H, Agarwal S, Gupta VK (2021) A critical review on the use of potentiometric based biosen-
sors for biomarkers detection. Biosens Bioelectron 184:113252
20. Karimi-Maleh H, Khataee A, Karimi F, Baghayeri M, Fu L, Rouhi J, Karaman C, Karaman
O, Boukherroub R (2021) A green and sensitive guanine-based DNA biosensor for idaru-
bicin anticancer monitoring in biological samples: a simple and fast strategy for control of
health quality in chemotherapy procedure confirmed by docking investigation. Chemosphere
291:132928. https://doi.org/10.1016/j.chemosphere.2021.132928
21. Karimi-Maleh H, Karimi F, Fu L, Sanati AL, Alizadeh M, Karaman C, Orooji Y (2022)
Cyanazine herbicide monitoring as a hazardous substance by a DNA nanostructure biosen-
sor. J Hazard Mater 423:127058. https://doi.org/10.1016/j.jhazmat.2021.127058
22. Karimi-Maleh H, Ayati A, Davoodi R, Tanhaei B, Karimi F, Malekmohammadi S, Orooji
Y, Fu L, Sillanpää M (2021) Recent advances in using of chitosan-based adsorbents for
removal of pharmaceutical contaminants: a review. J Clean Prod 291:125880. https://doi.
org/10.1016/j.jclepro.2021.125880
23. Tasheva K, Kosturkova G (2010) Rhodiola rosea in vitro cultures peculiarities. Sci
Publications Univ Agron Sci Vet Med 2010b:103–112
24. Erst A, Yakubov V (2019) Regenerative in vitro capacity of rare species Rhodiola rosea
L. from various habitats. Contemp Probl Ecol 12:368–376
25. Nadjafi F, Bannayan M, Tabrizi L, Rastgoo M (2006) Seed germination and dormancy break-
ing techniques for Ferula gummosa and Teucrium polium. J Arid Environ 64:542–547
26. Bian F, Su J, Liu W, Li S (2018) Dormancy release and germination of Taxus yunnanensis
seeds during wet sand storage. Sci Rep 8:3205. https://doi.org/10.1038/s41598-018-21469-9
27. Liu D, Yu HL, Li FL, Guo HH (2011) An analysis of dormancy and dormancy release in
Taxus chinensis var. mairei seeds. Seed Sci Technol 39:29–43. https://doi.org/10.15258/
sst.2011.39.1.04
28. Muñoz-Gutiérrez L, Vargas-Hernández JJ, López-Upton J, Soto-Hernández M (2009) Effect
of cutting age and substrate temperature on rooting of Taxus globosa. New For 38:187–196.
https://doi.org/10.1007/s11056-009-9139-6
29. Iszkuło G, Boratyński A (2004) Interaction between canopy tree species and European yew
Taxus baccata (Taxaceae). Pol J Ecol 52:523–531
30. Hong CP, Kim J, Lee J, Yoo S, Bae W, Geem KR, Yu J, Jang I, Jo IH, Cho H (2021)
Gibberellin signaling promotes the secondary growth of storage roots in Panax ginseng. Int
J Mol Sci 22:8694
31. Lee J-W, Jo I-H, Kim J-U, Hong C-E, Kim Y-C, Kim D-H, Park Y-D (2018) Improvement
of seed dehiscence and germination in ginseng by stratification, gibberellin, and/or kinetin
treatments. Hortic Environ Biotechnol 59:293–301
32. Kim J, Shin W-R, Kim Y-H, Shim D, Ryu H (2021) Functional characterization of gibberellin
signaling-related genes in Panax ginseng. J. Plant Biotechnol. 48:148–155
33. Bilinska E, Buchwald W, Krajewska-Patan A (2010) Biology of germination of medicinal
plant seeds. Part XXIIa. Seeds of Rhodiola kirilowii (Regel.) Maxim from Crassulaceae fam-
ily. Herba Pol 56:7–13
34. Wang F, Cai C, Wen B (2011) Study on the characteristics of seed dormancy and germination
of Rauvolfia yunnanensis (Apocynaceae). Plant Divers Resour 33:229–234
35. Dong S, Zhang Y, Chen N, Li C, Gao H (2011) Study on characteristics of seeds dormancy
and germination of parasitic plant Cynomorium songaricum Rupr. Med. Plant. 2:1–12
36. Chen QL, Guo Y, Jiang Y, Tu P (2016) Mechanism of fluridone-induced seed germination of
Cistanche tubulosa. Pak J Bot 48:971–976
1092 Y. Zheng et al.
37. Wang J, Baskin JM, Baskin CC, Liu G, Yang X, Huang Z (2017) Seed dormancy and ger-
mination of the medicinal holoparasitic plant Cistanche deserticola from the cold desert of
Northwest China. Plant Physiol Biochem 115:279–285
38. Wenmei F, Geer S (2002) The comparison of the dynamic changes of endogenous hormones
between Cynomorium songaricum Rupr. and Avena chinensis Metag. during the germination
of their seeds. Acta Sci Nat Univ Neimonggol Nat Sci Ed 33:304–308
39. Yang Y, Yi X, Peng M, Zhou Y (2012) Stable carbon and nitrogen isotope signatures of root-
holoparasitic Cynomorium songaricum and its hosts at the Tibetan plateau and the surround-
ing Gobi desert in China. Isot Environ Health Stud 48:483–493
40. Jiang Z-H, Tanaka T, Sakamoto M, Jiang T, Kouno I (2001) Studies on a medicinal para-
sitic plant: lignans from the stems of Cynomorium songaricum. Chem Pharm Bull (Tokyo)
49:1036–1038
41. Cui J-L, Vijayakumar V, Zhang G (2018) Partitioning of fungal endophyte assemblages
in root-parasitic plant Cynomorium songaricum and its host Nitraria tangutorum. Front
Microbiol 9:666
42. Badhwar R, Sharma H (1963) A note on the germination of Podophyllum seeds. Indian For
89:445–447
43. Nadeem M, Palni L, Purohit A, Pandey H, Nandi S (2000) Propagation and conservation
of Podophyllum hexandrum Royle: an important medicinal herb. Biol Conserv 92:121–129
44. Li Z, Qin G, He J, Cao X (2010) Comparative analysis of fatty acid composition in seed
kernel and coat of Paeonia rockii seeds. Seed 29:34–36
45. Zhang S, Tang H, Li W, Zhang R, He L (2020) Hypocotyl germination and cotyledon ana-
tomical structure of Paeonia rockii seeds by soaking with gibberellic acid and warm water.
Acta Bot Boreali-Occident Sin 40:442–453
46. Peng C, Wang P (1998) An analysis of the main elements in predicting the graft survival rate
of Eucommia ulmoides Oliv. J Hubei Inst Natl 6:24–26
47. Fan G (2008) A comparative study on the gratification and survival rates 40 clones of
Eucommia ulmoides. J Northwest For Univ 23:91–94
48. Zhou C, Tan M, Liu J, Cheng X (2019) Experimental study on the grafting and preservation
of the seed source of Magnolia officinalis. Jilin For Sci Technol 48:7–14
49. Su F, Lan J, Feng L (2012) Experimental study on Camptotheca acuminata Grafting. North
Hortic 19:62–65
50. Tang Z, Wu X (2008) Study on seedling technique for Chinese Torreya in Jingdezhen City.
Acta Agric Jiangxi 20:110–111
51. Yu M (2014) Grafting test of Taxus wallichiana var.mairei. J Fujian For Sci Technol 4:132–135
52. Chen Z, Zhang L (1998) Research on propagation technology of Eucommia cuttings. Chin
Herb Med 29:334–337
53. Mai M, Wang M, Shi D (2011) Study on twig cutting propagation techniques of Cercidiphyllum
japonicum. J. Fujian For. Sci. Technol. 38:102–106
54. Fu R, Huang Q, Liu A, Xu L, Huang X (2006) Studies on cutting propagation of Taxus chi-
nensis var. mairei VII. Comparison of photosynthesis physiological characteristics between
the seed seedlings and the cutting seedlings. Chin J Eco-Agric 14:62–63
55. Fu R, Sun X, Huang Q, Zhu J, Xu L (2005) Studies on the cutting propagation of Taxus
chinensis var.mairei.III .Effects of foliage fertilizer and transparency on growth of the cut
seedlings. Chin J Eco-Agric 13:175–177
56. Fu R, Xiaodong S, Qi H, Xueling H, Luxia X (2005) Studies on the cutting propagation of
Taxus chinensis var. mairei. IV. Effects of rooting powder on the rooting of the cutting. Chin
J Eco-Agric 13:178–180
57. Fu R, Zhu J, Huang Q, Lin Z, Huang J (2006) Studies on the cutting propagation of Taxus
chinensis var mairei V Occurrence and control of pests and diseases of the cuttage seedling
in greenhouse. Chin J Eco-Agric 14:193–195
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1093
58. Fu R, Zhu J, Liu S, Fang D, Ke Y (2006) Studies on cutting propagation of Taxus chinensis
var. mairei VII. Comparison of physiological metabolism between the seed seedlings and the
cutting seedlings. Chin J Eco-Agric 14:64–65
59. Fu S, Huang Q, Hu Z (2005) Studies on the cutting propagation of Taxus chinensis var.mairei
.I .Effects of medium, season and biological measures on the cutting propagation. Chin J
Eco-Agric 13:37–38
60. Chakraborty A, Bhattacharya D, Ghanta S, Chattopadhyay S (2010) An efficient protocol for
in vitro regeneration of Podophyllum hexandrum, a critically endangered medicinal plant.
Indian J Biotechnol 9:217–220
61. Rajesh M, Sivanandhan G, Jeyaraj M, Chackravarthy R, Manickavasagam M, Selvaraj N,
Ganapathi A (2014) An efficient in vitro system for somatic embryogenesis and podophyl-
lotoxin production in Podophyllum hexandrum Royle. Protoplasma 251:1231–1243
62. Du S, Xiang T, Song Y, Huang L, Sun Y, Han Y (2015) Transgenic hairy roots of Tetrastigma
hemsleyanum: induction, propagation, genetic characteristics and medicinal components.
Plant Cell Tissue Organ Cult PCTOC 122:373–382
63. Hu W, Zheng Y, Xia P, Liang Z (2021) The research progresses and future prospects
of Tetrastigma hemsleyanum Diels et Gilg: a valuable Chinese herbal medicine. J
Ethnopharmacol 271:113836
64. Peng X, Ji Q, Wu H, Li Y (2015) Slow-growth conservation and clonal fidelity of Tetrastigma
hemsleyanum microplants. Vitro Cell Dev Biol-Plant 51:463–470
65. Jiang W, Fu Y, Zhou X, Fu C (2011) High-frequency shoot regeneration of nodal explants
from Tetrastigma hemsleyanum Diels et Gilg: a valuable medicinal plant. Afr J Biotechnol
10:12177–12181
66. Xu L, Liu S, Bai Y, Ding H, Hu X, Wu X, Xu H, Zheng B (2018) Effects of light intensity
treatments on photosynthetic characteristics in Tetrastigma hemsleyanum. J Zhejiang AF
Univ 35:467–475
67. Zhou J, Zheng Y, Zhang J, Karimi-Maleh H, Xu Y, Zhou Q, Fu L, Wu W (2020) Characterization
of the electrochemical profiles of lycoris seeds for species identification and infrageneric
relationships. Anal Lett 53:2517–2528. https://doi.org/10.1080/00032719.2020.1746327
68. Wang Y, Pan B, Zhang M, Du X, Wu W, Fu L, Zhou W, Zheng Y (2020) Electrochemical
profile recording for pueraria variety identification. Anal Sci 36:20P079
69. Fu L, Su W, Chen F, Zhao S, Zhang H, Karimi-Maleh H, Yu A, Yu J, Lin C-T (2021) Early
sex determination of Ginkgo biloba based on the differences in the electrocatalytic perfor-
mance of extracted peroxidase. Bioelectrochemistry 140:107829. https://doi.org/10.1016/j.
bioelechem.2021.107829
70. Fu L, Wang A, Lyv F, Lai G, Zhang H, Yu J, Lin C-T, Yu A, Su W (2018) Electrochemical
antioxidant screening based on a chitosan hydrogel. Bioelectrochemistry 121:7–10. https://
doi.org/10.1016/j.bioelechem.2017.12.013
71. Fu L, Zheng Y, Zhang P, Zhang H, Wu M, Zhang H, Wang A, Su W, Chen F, Yu J, Cai
W, Lin C-T (2019) An electrochemical method for plant species determination and classi-
fication based on fingerprinting petal tissue. Bioelectrochemistry 129:199–205. https://doi.
org/10.1016/j.bioelechem.2019.06.001
72. Xu Y, Lu Y, Zhang P, Wang Y, Zheng Y, Fu L, Zhang H, Lin C-T, Yu A (2020) Infrageneric
phylogenetics investigation of Chimonanthus based on electroactive compound profiles.
Bioelectrochemistry 133:107455. https://doi.org/10.1016/j.bioelechem.2020.107455
73. Fan B, Wang Q, Wu W, Zhou Q, Li D, Xu Z, Fu L, Zhu J, Karimi-Maleh H, Lin C-T (2021)
Electrochemical fingerprint biosensor for natural indigo dye yielding plants analysis.
Biosensors 11:155. https://doi.org/10.3390/bios11050155
74. Fu L, Zhu J, Karimi-Maleh H (2021) An analytical method based on electrochemical sensor
for the assessment of insect infestation in flour. Biosensors 11:325. https://doi.org/10.3390/
bios11090325
1094 Y. Zheng et al.
75. Zheng Y, Wang D, Li X, Wang Z, Zhou Q, Fu L, Yin Y, Creech D (2021) Biometric iden-
tification of Taxodium spp. and their hybrid progenies by electrochemical fingerprints.
Biosensors 11:403
76. Tikendra L, Potshangbam AM, Dey A, Devi TR, Sahoo MR, Nongdam P (2021) RAPD,
ISSR, and SCoT markers based genetic stability assessment of micropropagated Dendrobium
fimbriatum Lindl. Var. oculatum Hk. f.-an important endangered orchid. Physiol Mol Biol
Plants 27:341–357
77. Roy J, Banerjee N (2003) Induction of callus and plant regeneration from shoot-tip explants
of Dendrobium fimbriatum Lindl. var. oculatum Hk. f. Sci Hortic 97:333–340
78. Da Silva JAT, Tsavkelova EA, Ng TB, Parthibhan S, Dobránszki J, Cardoso JC, Rao M, Zeng
S (2015) Asymbiotic in vitro seed propagation of Dendrobium. Plant Cell Rep 34:1685–1706
79. Luo J, Cheng Z, Long C (2006) Studies on the rapid propagation and in vitro storage of
Dendrobium candidum. Guihaia 26:69–73
80. Zhang A, Wang H, Shao Q, Xu M, Zhang W, Li M (2015) Large scale in vitro propagation of
Anoectochilus roxburghii for commercial application: pharmaceutically important and orna-
mental plant. Ind Crop Prod 70:158–162
81. Gam DT, Khoi PH, Ngoc PB, Linh LK, Hung NK, Anh PTL, Thu NT, Hien NTT, Khanh TD,
Ha CH (2020) LED lights promote growth and flavonoid accumulation of Anoectochilus rox-
burghii and are linked to the enhanced expression of several related genes. Plan Theory 9:1344
82. Lin X, Jiang X, Yang Z, Ma X, Yan X, Yang L (2018) Tissue culture rapid propagation and
transplantation techniques of Anoectochilus roxburghii (Wall) Lind. Med Plant 9:43–46
83. Ket N, Hahn E, Park S, Chakrabarty D, Paek K (2004) Micropropagation of an endangered
orchid Anoectochilus formosanus. Biol Plant 48:339–344
84. Zhang F, Lv Y, Dong H, Guo S (2010) Analysis of genetic stability through Intersimple
sequence repeats molecular markers in micropropagated plantlets of Anoectochilus formosa-
nus H AYATA, a medicinal plant. Biol Pharm Bull 33:384–388
85. Du X, Sun N, Hayashi J, Chen Y, Sugiura M, Shoyama Y (2003) Hepatoprotective and anti-
hyperliposis activities of in vitro cultured Anoectochilus formosanus. Phytother Res Int J
Devoted Pharmacol Toxicol Eval Nat Prod Deriv 17:30–33
86. Lv T, Teng R, Shao Q, Wang H, Zhang W, Li M, Zhang L (2015) DNA barcodes for the iden-
tification of Anoectochilus roxburghii and its adulterants. Planta 242:1167–1174
87. Dan Y, Yu X, Guo S-X, Meng Z (2012) Effects of forty-two strains of orchid mycorrhizal
fungi on growth of plantlets of Anoectochilus roxburghii. Afr J Microbiol Res 6:1411–1416
88. Ma J, Rong K, Cheng K (2012) Research and practice on biodiversity in situ conservation in
China: progress and prospect. Biodivers Sci 20:551–558
89. Heywood VH (2015) In situ conservation of plant species–an unattainable goal? Isr J Plant
Sci 63:211–231
90. Tan S, Zhong Y, Yang F, Gong X (2021) The impact of Nanshan National Park concession
policy on farmers’ income in China. Glob Ecol Conserv 31:e01804
91. Wu J, Wu G, Zheng T, Zhang X, Zhou K (2020) Value capture mechanisms, transaction
costs, and heritage conservation: a case study of Sanjiangyuan National Park. China Land
Use Policy 90:104246
92. Wang G, Innes JL, Wu SW, Krzyzanowski J, Yin Y, Dai S, Zhang X, Liu S (2012) National
park development in China: conservation or commercialization? Ambio 41:247–261
93. He S, Su Y, Wang L, Gallagher L, Cheng H (2018) Taking an ecosystem services approach for
a new national park system in China. Resour Conserv Recycl 137:136–144
94. Peng H, Zhang J, Lu L, Tang G, Yan B, Xiao X, Han Y (2017) Eco-efficiency and its determi-
nants at a tourism destination: a case study of Huangshan National Park. China Tour Manag
60:201–211
95. Volis S, Blecher M (2010) Quasi in situ: a bridge between ex situ and in situ conservation of
plants. Biodivers Conserv 19:2441–2454
96. Barazani O, Perevolotsky A, Hadas R (2008) A problem of the rich: prioritizing local plant
genetic resources for ex situ conservation in Israel. Biol Conserv 141:596–600
Pharmacological Properties and Tissue Culture Method of Endangered Medicinal Plants 1095
97. Perrino EV, Wagensommer RP (2021) Crop wild relatives (CWR) priority in Italy: distribu-
tion, ecology, in situ and ex situ conservation and expected actions. Sustainability 13:1682
98. Patel AK, Lodha D, Shekhawat NS (2020) An improved micropropagation protocol for the
ex situ conservation of Mitragyna parvifolia (Roxb.) Korth.(Rubiaceae): an endangered tree
of pharmaceutical importance. Vitro Cell. Dev. Biol.-Plant. 56:817–826
99. Grigoriadou K, Sarropoulou V, Krigas N, Maloupa E, Tsoktouridis G (2020) GIS-facilitated
effective propagation protocols of the endangered local endemic of crete Carlina diae (Rech.
f.) Meusel and A. Kástner (Asteraceae): serving ex situ conservation needs and its future
sustainable utilization as an ornamental. Plan Theory 9:1465
100. Thapa BB, Thakuri LS, Joshi PR, Chand K, Rajbahak S, Sah AK, Shrestha R, Paudel MR,
Park SY, Pant B (2020) Ex-situ conservation and cytotoxic activity assessment of native
medicinal orchid: Coelogyne stricta. J Plant Biotechnol 47:330–336
101. Wei X, Jiang M (2021) Meta-analysis of genetic representativeness of plant populations
under ex situ conservation in contrast to wild source populations. Conserv Biol 35:12–23
102. Ciocarlan N, Izverscaia T, Ghendov V (2018) Spontaneous medicinal plants research and
ex-situ conservation in the national botanical garden (institute), Republic of Moldova. Rev
Bot 16:50–56
103. Streczynski R, Clark H, Whelehan LM, Ang S-T, Hardstaff LK, Funnekotter B, Bunn E,
Offord CA, Sommerville KD, Mancera RL (2019) Current issues in plant cryopreserva-
tion and importance for ex situ conservation of threatened Australian native species. Aust J
Bot 67:1–15
104. Engels JM, Ebert AW (2021) A critical review of the current global ex situ conservation
system for plant agrobiodiversity. I. History of the development of the global system in
the context of the political/legal framework and its major conservation components. Plan
Theory 10:1557
105. Dostatny DF, Żurek G, Kapler A, Podyma W (2021) The ex situ conservation and potential
usage of crop wild relatives in Poland on the example of grasses. Agronomy 11:94
Natural Compounds with Pharmacological
Properties in Clinical Trials
1 Introduction
Natural products serve as a repository for identifying novel leads that may be uti-
lized to treat various illnesses, including cancer, inflammation, and liver disease.
Natural chemicals or their derivatives have been the source of more than 50% of all
pharmaceutical drugs found to date [1]. Nowadays, medicinal plants have received
increased interest in both studies and clinical practice due to their minimal side
effects and availability. In most cases, herbal medications are utilized as a comple-
mentary and adjunct treatment for various conditions, such as type 2 diabetes mel-
litus (T2DM) or cardiovascular disease (CVD) [2]. This chapter introduces some
common types of these natural compounds and reviews clinical trials performed on
these natural compounds. The possible mechanisms for the observed impact are
then described.
2 Curcumin
(Gpx) enzyme activity. It was also associated with beneficial potencies in hyperan-
drogenemia (by lowering Dehydroepiandrosterone) and hyperglycemia (by lower-
ing fasting plasma glucose) in these patients [24, 25]. Curcumin supplementation
with a lower dose, 500 mg per day for 12 weeks, resulted in weight loss, body mass
index (BMI) reduction, and glycemic control properties by reducing fasting glu-
cose, insulin, insulin resistance, and enhancing insulin sensitivity. After the inter-
vention, it was also associated with lower total cholesterol, low-density
lipoprotein-cholesterol (LDL-chol), total cholesterol (total-chol) to high-density
lipoprotein-cholesterol (HDL-chol) ratio, and higher HDL-chol levels. Besides,
gene expression of peroxisome proliferator-activated receptor gamma (PPAR-γ)
and LDL receptor increased [26]. Glycemic control properties of curcumin by
reducing serum insulin are shown in another trial in a dose of 1000 mg per day
divided into two equal doses for 6 weeks [27].
Seven-week curcumin supplementation in the form of 80 mg nano-micelle cur-
cumin capsules two times per day may be helpful to prevent and even treat oral
mucositis severity and pain, a side effect of chemotherapy or radiotherapy in patients
with cancer who underwent chemotherapy with or without head and neck radio-
therapy [28].
In hemodialysis, which is associated with higher oxidative stress and inflamma-
tion, supplementation with 1 g/day curcumin for 12 weeks resulted in higher cata-
lase activity and preserved glutathione peroxidase activity. However, it did not
change malondialdehyde (MDA) and high-sensitive C-reactive protein (hs-CRP)
concentrations and glutathione reductase activity [29]. In a similar study in hemodi-
alysis patients, the anti-inflammatory of nano-curcumin supplementation for 3
months has been shown by lowering gene expression and concentrations of interleu-
kin 6 (IL-6) and TNF-α [30]. In kidney disease conditions, Gut dysbiosis and ure-
mic toxins production are also prevalent and associated with inflammation.
Curcumin supplementation in juice containing 2.5 gr turmeric three times weekly
for 3 months showed modulating effects by lowering p-CS, a uremic toxin [31].
In the intensive care unit (ICU) admitted critically ill patients with traumatic
brain injury, daily intake of 500 mg curcuminoids for seven days resulted in a sig-
nificant reduction of inflammatory markers IL-6, TNF-α, monocyte chemoattractant
protein-1 (MCP-1), and C-reactive protein (CRP) [32].
Intake of 400 mg per day curcumagalactomannosides for 6 weeks in individuals
with osteoarthritis had positive effects on pain and osteoarthritis symptoms com-
pared to glucosamine hydrochloride and chondroitin sulfate mixture [33].
Curcumin‘s anti-inflammatory benefits have been linked to a number of different
mechanisms, one of which is the inhibition of the nuclear factor kappa-light-chain-
enhancer of activated B cells (NF-kb) [34].
Curcumin may affect lipid profile by inhibiting lipid synthesis via downregula-
tion of key enzymes, including β-hydroxy β-methylglutaryl-CoA (HMG-CoA)
reductase, sterol regulatory element-binding protein-1/2 (SREBP-1/2), and fatty
acid synthase (FAS) [35–37]. Curcumin enhances cholesterol 7alpha-hydroxylase
(CYP7a1) gene expression resulting in biliary lipid excretion [37]. Curcumin stimu-
lates the PPAR-γ activation [38]. Besides, by activating AMP-activated protein
1100 M. Noormohammadi and F. Shidfar
kinase (AMPK), curcumin stimulates lipid catabolism and regulates energy homeo-
stasis in cells [39]. By increasing lipoprotein lipase activity, curcumin reduces tri-
glyceride levels and dyslipidemia [40]. Also, curcumin reduces the amount of free
fatty acids in the blood, influences the absorption and transportation of cholesterol
through the digestive tract, and reduces the risk of lipid peroxidation [41]. As a
consequence of curcumin‘s ability to enhance the hemodynamic state of arteries
and boost nitric oxide synthase, it may reduce blood pressure [42].
Glycemic control properties of curcumin are shown to be via PPAR-γ activation,
increasing some glycolysis pathway enzyme activity (including hepatic glucoki-
nase) and decreasing some enzymes gluconeogenesis pathway activity (including
glucose-6-phosphatase and phosphoenolpyruvate carboxykinase) and also glucose
uptake stimulation by elevating gene expression of glucose transporter [20, 43, 44].
AMPK phosphorylation and activation as another role of curcumin on glucose
uptake increase. By enhancing mitogen-activated protein kinase kinase
(MEK)3/6-p38 and mitogen-activated protein kinase (MAPK) signaling pathways,
curcumin improves cellular glucose uptake by cells [45, 46]. Furthermore, curcumin
has been shown to ameliorate insulin resistance by boosting adiponectin levels and
reducing leptin levels [43, 44]. Besides, curcumin increases the production of
glucagon-like peptide-1 (GLP-1) [47]. Additionally, curcumin may increase insulin
secretion by increasing B-cell activity [48]. Curcumin antioxidant properties are
another mechanism for reducing glucose intolerance [49].
3 Ginger
4 Sumac
Rhus coriaria L., often known as “sumac,” is a herb from the family Anacardiaceae
and the genus Rhus. Sumac has been used as a spice and herb throughout many
countries. A variety of biological activities, including antioxidant, hypoglycemic,
anti-inflammatory, cytotoxic, and anti-thrombin activity, are attributed to sumac.
Sumac‘s biological capabilities are attributed to the existence of several bioactive
Phyto-components such as phenolic acids (gallic acid) and flavonoids, which are
present in high concentrations (quercetin, kaempferol) [62].
Sumac‘s glycemic control properties have been documented in various in vitro
and in vivo animal and human investigations. Several polyphenols in sumac contain
insulin-like characteristics, and they have been shown to be favorable for enhancing
1102 M. Noormohammadi and F. Shidfar
insulin sensitivity and decreasing oxidative stress [62]. supplementation with sumac
powder 3 g/day after 3 months resulted in a significant decrease in serum glucose,
HbA1c, apoB, apoB/apoA-I ratio, and a significant increase in apoA-I and TAC In
T2DM patients [63]. In T2DM patients, 3 g/day sumac powder intake for 3 months
has shown a significant enhancement for PON1 activity and a significant decrease
in serum glucose, insulin, homeostatic model assessment of insulin resistance
(HOMA-IR), hs-CRP, and MDA [64].
In patients with NAFLD, sumac supplementation in combination with a low-
calorie diet has shown benefits for liver function. In a 12-week supplementation
with 2 gr/day sumac, a trial has shown significantly lower hepatic fibrosis, alanine
transaminase (ALT), AST, fasting plasma glucose, serum insulin, HbA1c,
HOMA-IR, MDA, and hs-CRP, in the intervention group rather than the pla-
cebo [65].
The combination of sumac supplementation and a restricted-calorie diet has
shown to be favorable for obesity control. Restricted-calorie diet along with daily
intake of 3000 mg sumac in overweight or obese adult women with depression for
12 weeks was superior to restricted-calorie diet alone on weight loss, and anthropo-
metric measurements decrease including BMI, body fat, visceral fat, waist, and hip
circumference and MDA levels, an oxidative stress marker [66].
Sumac may have cardioprotective properties in patients with dyslipidemia. Four-
week daily consumption of 1 gr sumac in patients with elevated total-chol and/or
triglycerides resulted in lower BMI, systolic blood pressure (SBP), diastolic blood
pressure (DBP), and total-chol [67]. In a similar study on patients with dyslipid-
emia, sumac supplementation for 6 weeks resulted in elevated HDL-chol and apo-
lipoprotein A-1 [68].
Since no changes have been shown for insulin and GLUT-4 gene expression,
Rhus coriaria L. may act in its glycemic control effect by modulating insulin secre-
tion or function [69]. According to in vitro studies, Rhus coriaria L. may inhibit
α-amylase [70]. Phenolic acids and flavonoids, such as gallic acid, methyl gallate,
kaempferol, and quercetin in Rhus coriaria L., may have potencies for glycemic
control [71, 72]. Quercetin has also shown a favorable effect on glycemic control
and total-chol and LDL-chol lowering properties [73]. Flavonoids may enhance the
preservation of β-cell by antioxidant effects [74]. Rhus coriaria L. has several ingre-
dients with antioxidant activity, including tannin and gallic acids that are helpful in
ROS scavenging [75].
5 Lemon Balm
Melissa officinalis (MO), often known as lemon balm, is a plant from the mint fam-
ily Lamiaceae. MO is a rich source of active phytochemicals such as triterpenes,
flavonols, and phenolic acid, among other things. MO has been used as a remedy in
Asian traditional medicine, including rheumatoid arthritis, gastrointestinal
Natural Compounds with Pharmacological Properties in Clinical Trials 1103
problems, and neurological disorders, among others, even though several primary
studies are looking at the effects of MO in chronic cardiometabolic illnesses [2].
Melissa officinalis may have beneficial properties in lipid profile, glycemic con-
trol, and inflammation in T2DM. Supplementation with 700 mg/d Melissa officina-
lis for 12 weeks in patients with T2DM resulted in a significant lower fasting
glucose, HbA1c, triglycerides, hs-CRP, and SBP and a significant increase in beta
cell activity, HDL-chol [76]. It also had favorable effects in apo-lipoprotein A-I,
apo-lipoprotein B/ apo-lipoprotein A-I in patients with T2DM but did not show any
positive properties on intercellular adhesion molecule-1 (ICAM-1), AST and, alka-
line phosphatase (ALP) [77]. The toxic effects of M. officinalis extract on liver tis-
sue were shown at very high doses (0.450 and 1.350 g/kg). According to the findings
of this research, using 700 mg/day of a hydroalcoholic extract of M. officinalis in
individuals with T2DM is considered safe [77].
In patients with chronic stable angina, 3000 mg daily intake of Melissa officina-
lis for 8 weeks reduced triglycerides, total-chol, LDL-chol, MDA, and hs-CRP and
enhanced PON1 and HDL-chol [78].
Melissa officinalis has shown to be effective in lowering SBP and DBP [79]. It
may also effectively lower serum triglycerides in patients with T2DM and dyslipid-
emia [80] and LDL-chol and AST in individuals with borderline hyperlipid-
emia [81].
M. officinalis has been shown to perform its antidiabetic roles by enhancing glu-
cokinase activity and gene expression and reducing glucose-6-phosphatase and
phosphoenolpyruvate carboxykinase [82].
HMG-CoA reductase, sterol regulatory element-binding protein-1c (SREBP-1c),
acetyl CoA carboxylase 1 (ACC1), stearoyl CoA desaturase 1 (SCD1), and FAS are
among the genes that have been shown to be downregulated by M. officinalis. These
genes are involved in fatty acid production and may be responsible for the plant‘s
antihyperlipidemic actions [83]. M. officinalis is shown to activate the AMPK/ACC
pathway resulting in glucose control regulation and insulin resistance reduction in
adipocytes besides inhibition of fat accumulation in adipocyte tissue [84].
6 Cinnamon
Cinnamon is a plant related to the Lauraceae family [85]. It is a popular spice that
many civilizations around the globe have utilized for hundreds of years. In addition
to its culinary applications, cinnamon is considered a cure for respiratory, digestive,
and gynecological disorders in traditional Ayurveda medicine. It is possible to
employ almost every component of the cinnamon tree for medicinal or culinary
purposes, such as the bark, leaves, flowers, fruits, and roots. All sections of the plant
contain the same array of hydrocarbons in variable quantities, with primary ele-
ments such as cinnamaldehyde (bark), eugenol (leaf), and camphor (seed) being
found in varying amounts (root). Cinnamon has a broad range of therapeutic effects,
and this chemical diversity is probably responsible for the extensive range of
1104 M. Noormohammadi and F. Shidfar
benefits seen [86]. According to studies, cinnamon has favorable properties on gly-
cemic control, blood pressure, weight control, and lipid profile [85]. The active
component cinnamaldehyde has been proposed, which has been linked to cinna-
mon‘s blood glucose-lowering properties [87].
Several studies have examined the cinnamon effect on patients with diabetes.
Supplementation with 1000 mg cinnamon for 3 months in those with T2DM reduced
anthropometric indices including BMI, body fat, and visceral fat, had glycemic con-
trol properties by reducing fasting glucose, 2-hour postprandial glucose, HbA1C,
fasting insulin, and insulin resistance, and enhanced lipid profile including total-
chol, LDL-chol, and HDL-chol especially in those with basal BMI ≥ 27 [88]. In
contrast, 3 gr of cinnamon consumption per day for 8 weeks did not significantly
affect glycemic control, advanced glycation end products (AGE), and antioxidant
status in adults with T2DM [89]. In adults with T2DM, 3000 mg cinnamon for 8
weeks did not enhance hs-CRP, TNF-α, IL-6, NF-kb, Sirtuin-1, and vascular adhe-
sion molecules, ICAM-1 and vascular cell adhesion molecule 1 (VCAM-1) [90,
91]. To compare different doses of cinnamon, another study showed that daily con-
sumption of 1, 3, or 6 g of cinnamon reduced serum glucose, triglyceride, LDL-
chol, and total-chol in people with T2DM [92].
In patients with metabolic syndrome, daily 3 gr intake of cinnamon for 16 weeks
improved fasting blood glucose, glycosylated hemoglobin, blood pressure, anthro-
pometric indices, and lipid profile [93].
Supplementation with 1500 mg cinnamon in 3 equally divided doses in over-
weight or obese women with PCOS for 8 weeks led to weight loss, had glucose-
lowering properties, and also decreased insulin and HOMA-IR. Besides,
supplementation reduced lipid markers, including total-chol and LDL-chol but
HDL-chol increased [94]. In a similar study, 1500 mg cinnamon in 3 equal divided
doses in women with PCOS for 12 weeks resulted in reduced fasting insulin,
HOMA-IR, and LDL [95].
In patients with NAFLD, daily consumption of 1.5 gr cinnamon for 12 weeks led
to a significant reduction in HOMA-IR, fasting glucose, total-chol, triglyceride,
ALT, AST, gamma glutamine transpeptidase (GGT), and hs-CRP. However, HDL
levels did not change [96].
In patients with migraine, daily consumption of 1800 mg cinnamon divided into
three equal doses for 2 months resulted in lower IL-6 and NO and reduced the fre-
quency, severity, and duration of headache [97].
Cinnamon contains anti-inflammatory effects [86, 98]. It is claimed that cinna-
mon contains certain narcotic, opioid, and anti-inflammatory substances such as
cinnamaldehyde, eugenol, and terpene. The anti-inflammatory feature has been
reported by cinnamaldehyde [99]. Cinnamaldehyde, the primary bioactive compo-
nent of cinnamon, may inhibit the synthesis of inflammatory cytokines such as IL-6,
IL-1beta, and TNF-α by inhibiting the expression of cyclooxygenase and nitric
oxide synthase [98, 100–103]. Several of cinnamon‘s constituents and metabolites
have been shown to modulate the release of inflammatory mediators [104, 105]. It
seems that cinnamon decreases NO’s metabolites, such as superoxide and peroxyni-
trite, which results in a reduction in the inflammation induced by NO, according to
Natural Compounds with Pharmacological Properties in Clinical Trials 1105
the research [98, 106, 107]. Terpene ingredients in cinnamon have been shown to
inhibit arachidonic acid metabolism and nitric oxide synthase enzyme [108].
Besides, Eugenol has been shown to be a potent inhibitor of arachidonic acid metab-
olism and antihistamine function resulting in the prohibition of inflammation [109].
Cinnamon may act as a scavenge for reactive carbonyl species resulting in the inhi-
bition of the generation of advanced glycation end products [110].
Cinnamon has been shown to have various benefits, including decreasing stom-
ach emptying, decreasing insulin levels, and increasing GLP-1 [111]. Cinnamon
extracts have shown to activate glycogen synthase, increase glucose absorption, and
inhibit glycogen synthase kinase-3 [112, 113]. Cinnamon extracts also stimulated
insulin receptor kinase and blocked dephosphorylation of the insulin receptor,
resulting in the insulin receptor being phosphorylated to its maximum level [113].
According to in vitro studies, stimulation of the insulin receptor by a water-soluble
polyphenol Type A polymer derived from cinnamon has been found to increase
insulin action by activating the insulin receptor. Methyl hydroxy chalcone polymer,
derived from cinnamon, is hypothesized to be responsible for a variety of biological
actions, including the reduction of insulin resistance [113–115].
Cinnamate, a phenolic substance found in cinnamon, has been shown to reduce
lipid peroxidation by increasing the activity of antioxidant enzymes in the liver [116].
7 Rheum Ribes
Rheum ribes is a plant that grows in the subtropical and temperate regions of the
world and is a member of the Polygonaceae family. Many active phytochemicals
and crude medicines may be found in this plant native to Asian nations. Flavonoids
(quercetin and catechin), anthocyanins (cyaniding), and stilbenes are among the
biologically active phytochemicals. The ethnomedicinal properties of Rheum ribes
include laxative, antioxidative, analgesic, antidiabetic, anti-mutagenic, cathartic,
anti-inflammatory, anticancer, antibacterial, hepatoprotective, antiplatelet, diarrhea,
measles, cholagogue, stomachic, smallpox, hemorrhoids, antiemetic, and antipsori-
atic properties [117].
Rheum ribes has been shown to have positive effects in diabetes conditions.
Following 6-week supplementation with Rheum ribes on extract improved insulin
sensitivity and oxidative markers, including malondialdehyde and high sensitivity
CRP in patients with T2DM [118].
Rheum ribes in the form of syrup may be helpful as a remedy on children with
Shigella caused dysentery diarrhea [119].
According to one study, Rheum ribes may have anti-depressive properties, which
observed this effect after a 6-week daily intake of 1200 mg Rheum ribes hydroalco-
holic extract in patients with major depressive disorder [120].
In diabetes conditions, Rheum ribes stalk extract has shown positive impacts on
serum lipid and glucose. Daily intake of 1200 mg Rheum ribes reduced fasting
1106 M. Noormohammadi and F. Shidfar
blood glucose, LDL-chol, and total-chol in patients with T2DM with hypercholes-
terolemia [121].
According to in vitro research, Rheum ribes might affect insulin secretion;
Because Ca2+ plays an important role in insulin exocytosis, the Rheum ribes extract
may function by elevating cytoplasmic Ca2+ via boosting the flow of extracellular
Ca2+ and intracellular Ca2+ [122].
The stem of the Rheum ribes contains large amounts of flavonoids, including
quercetin [121]. In addition, the rhizome of Rheum ribes contains stilbenes with
antioxidant activity [123]. Antioxidants effectively reduce oxidative stress disorders
by removing ROS [121]. Studies have indicated that aloe-emodin is the bioactive
component of Rheum ribes that significantly impacts the inflammatory state, most
likely via a decrease in pro-inflammatory cytokine production [124].
8 Urtica Dioica
Urtica dioica (Nettle), a member of the Urticaceae family, is well known for its
wide variety of biological activities and favorable benefits to human health. In addi-
tion to flavonoids, silicic acid, and butyric acid, potassium ions, nitrates and volatile
oil, histamine and serotonin, acetylcholine, formic acid, and leukotriene. This
plant’s anti-inflammatory, anti-hyperglycemic, anti-proliferative, immunomodula-
tory, diuretic, antiplatelet aggregation, anti-allergic, antimicrobial, antioxidant, anti-
ulcer, and analgesic properties are among its many therapeutic benefits [125].
Urtica dioica leaf extract has inflammatory lowering properties in patients with
inflammatory bowel disease. Twelve-week daily supplementation with 1200 mg
Urtica dioica leaf extracts significantly reduced TNF-α and fecal calprotectin [126].
It also effectively lowers hs-CRP and platelet count and enhances antioxidant
defense by increasing superoxide dismutase enzyme (SOD) activity in these
patients [127].
Urtica dioica may have significant disease-managing results in men with benign
prostatic hyperplasia. According to a recent study, 12-week daily consumption of
450 mg Urtica dioica root extract has significant positive impacts on International
Prostate Symptoms Score, hs-CRP, and MDA and SOD activity [128].
In diabetes conditions, Urtica dioica has shown oxidative stress-lowering prop-
erties. Eight-week daily intake of 100 mg hydroalcoholic extract of Urtica dioica
significantly decreased IL-6 and hs-CRP [129] and increased TAC and SOD in
patients with T2DM [130]. Three-month supplementation with Urtica dioica leaf
extract in 500 mg capsule every 8 hours combined with anti-hyperglycemic medi-
cine significantly reduced fasting glucose, 2 hours postprandial glucose, and
HbA1c [131].
Several in vivo researches have looked at how Urtica dioica might influence
inflammatory and antioxidant markers in the body. CRP is generated in reaction to
Natural Compounds with Pharmacological Properties in Clinical Trials 1107
9 Conclusions
Nowadays, the consumption of several natural compounds and plants has increased
widely due to the observed health benefits. Cell culture, animal, and human studies
have introduced several natural compounds with extensive effects on various condi-
tions, including malignancies, infections, cardiometabolic risk factors like stroke,
myocardial infarction, dyslipidemia, insulin resistance, diabetes mellitus type 2,
and even weight management. However, it is essential to be sure of safety aspects
such as the physiological dose. It is necessary to receive these compounds in dosage
to observe the desired effects without side effects. The mechanisms by which these
compounds exert health-promoting effects have not been clearly understood and
studied. There is also a need for further studies to provide conclusive evidence of
the side effects. Besides, the safety of consuming these compounds in certain condi-
tions and diseases should also be noted. Considering all these conditions, it seems
that natural compounds can be used to provide health-supporting properties.
1108 M. Noormohammadi and F. Shidfar
References
1. Clark AM (1996) Natural products as a resource for new drugs. Pharm Res 13(8):1133–1141
2. Heshmati J, Morvaridzadeh M, Sepidarkish M, Fazelian S, Rahimlou M, Omidi A et al
(2020) Effects of Melissa officinalis (Lemon Balm) on cardio-metabolic outcomes: a system-
atic review and meta-analysis. Phytother Res 34(12):3113–3123
3. Lestari ML, Indrayanto G (2014) Curcumin. Profiles Drug Subst Excip Relat Methodol
39:113–204. https://doi.org/10.1016/b978-0-12-800173-8.00003-9
4. Gupta SC, Kismali G, Aggarwal BB (2013) Curcumin, a component of turmeric: from farm
to pharmacy. Biofactors 39(1):2–13
5. Jabczyk M, Nowak J, Hudzik B, Zubelewicz-Szkodzińska B (2021) Curcumin in metabolic
health and disease. Nutrients 13(12):4440
6. Meeting JFWECoFA, Organization WH (2013) Evaluation of certain food additives and con-
taminants: seventy-seventh report of the joint FAO/WHO expert committee on food addi-
tives. World Health Organization
7. Alm-Eldeen AA, Mona MH, Shati AA, El-Mekkawy HI (2015) Synergistic effect of black
tea and curcumin in improving the hepatotoxicity induced by aflatoxin B1 in rats. Toxicol Ind
Health 31(12):1269–1280. https://doi.org/10.1177/0748233713491807
8. Dattani JJ, Rajput DK, Moid N, Highland HN, George LB, Desai KR (2010) Ameliorative
effect of curcumin on hepatotoxicity induced by chloroquine phosphate. Environ Toxicol
Pharmacol 30(2):103–109. https://doi.org/10.1016/j.etap.2010.04.001
9. Coban D, Milenkovic D, Chanet A, Khallou-Laschet J, Sabbe L, Palagani A et al (2012)
Dietary curcumin inhibits atherosclerosis by affecting the expression of genes involved in
leukocyte adhesion and transendothelial migration. Mol Nutr Food Res 56(8):1270–1281.
https://doi.org/10.1002/mnfr.201100818
10. Leclercq IA, Farrell GC, Sempoux C, dela Peña A, Horsmans Y. (2004) Curcumin inhibits
NF-kappaB activation and reduces the severity of experimental steatohepatitis in mice. J
Hepatol 41(6):926–934. https://doi.org/10.1016/j.jhep.2004.08.010
11. Kelany ME, Hakami TM, Omar AH (2017) Curcumin improves the metabolic syndrome
in high-fructose-diet-fed rats: role of TNF-α, NF-κB, and oxidative stress. Can J Physiol
Pharmacol 95(2):140–150. https://doi.org/10.1139/cjpp-2016-0152
12. Maithilikarpagaselvi N, Sridhar MG, Swaminathan RP, Zachariah B (2016) Curcumin pre-
vents inflammatory response, oxidative stress and insulin resistance in high fructose fed male
Wistar rats: potential role of serine kinases. Chem Biol Interact 244:187–194. https://doi.
org/10.1016/j.cbi.2015.12.012
13. Ramirez-Tortosa MC, Ramirez-Tortosa CL, Mesa MD, Granados S, Gil A, Quiles JL
(2009) Curcumin ameliorates rabbits’s steatohepatitis via respiratory chain, oxidative
stress, and TNF-alpha. Free Radic Biol Med 47(7):924–931. https://doi.org/10.1016/j.
freeradbiomed.2009.06.015
14. Cho JW, Lee KS, Kim CW (2007) Curcumin attenuates the expression of IL-1beta, IL-6, and
TNF-alpha as well as cyclin E in TNF-alpha-treated HaCaT cells; NF-kappaB and MAPKs
as potential upstream targets. Int J Mol Med 19(3):469–474
15. Sidhu GS, Singh AK, Thaloor D, Banaudha KK, Patnaik GK, Srimal RC et al (1998)
Enhancement of wound healing by curcumin in animals. Wound Repair Regen 6(2):167–177
16. Farzaei MH, Zobeiri M, Parvizi F, El-Senduny FF, Marmouzi I, Coy-Barrera E et al (2018)
Curcumin in liver diseases: a systematic review of the cellular mechanisms of oxidative stress
and clinical perspective. Nutrients 10(7):855
17. Saadati S, Hatami B, Yari Z, Shahrbaf MA, Eghtesad S, Mansour A et al (2019) The effects
of curcumin supplementation on liver enzymes, lipid profile, glucose homeostasis, and
hepatic steatosis and fibrosis in patients with non-alcoholic fatty liver disease. Eur J Clin
Nutr 73(3):441–449. https://doi.org/10.1038/s41430-018-0382-9
Natural Compounds with Pharmacological Properties in Clinical Trials 1109
18. Saadati S, Sadeghi A, Mansour A, Yari Z, Poustchi H, Hedayati M et al (2019) Curcumin and
inflammation in non-alcoholic fatty liver disease: a randomized, placebo controlled clinical
trial. BMC Gastroenterol 19(1):133. https://doi.org/10.1186/s12876-019-1055-4
19. Mirhafez SR, Azimi-Nezhad M, Dehabeh M, Hariri M, Naderan RD, Movahedi A et al
(2021) The effect of curcumin phytosome on the treatment of patients with non-alcoholic
fatty liver disease: a double-blind, randomized, placebo-controlled trial. Adv Exp Med Biol
1308:25–35. https://doi.org/10.1007/978-3-030-64872-5_3
20. Zhang D-w, Fu M, Gao S-H, Liu J-L. Curcumin and diabetes: a systematic review. Evid
Based Complement Alternat Med 2013;2013
21. Thota RN, Rosato JI, Dias CB, Burrows TL, Martins RN, Garg ML (2020) Dietary supple-
mentation with curcumin reduce circulating levels of glycogen synthase kinase-3β and islet
amyloid polypeptide in adults with high risk of type 2 diabetes and Alzheimer’s disease.
Nutrients 12(4). https://doi.org/10.3390/nu12041032
22. Bateni Z, Rahimi HR, Hedayati M, Afsharian S, Goudarzi R, Sohrab G (2021) The effects of
nano-curcumin supplementation on glycemic control, blood pressure, lipid profile, and insu-
lin resistance in patients with the metabolic syndrome: a randomized, double-blind clinical
trial. Phytother Res 35(7):3945–3953. https://doi.org/10.1002/ptr.7109
23. Mohammadi M (2019) Oxidative stress and polycystic ovary syndrome: a brief review. Int J
Prev Med 10:86
24. Heshmati J, Moini A, Sepidarkish M, Morvaridzadeh M, Salehi M, Palmowski A et al (2021)
Effects of curcumin supplementation on blood glucose, insulin resistance and androgens in
patients with polycystic ovary syndrome: a randomized double-blind placebo-controlled clin-
ical trial. Phytomedicine 80:153395. https://doi.org/10.1016/j.phymed.2020.153395
25. Heshmati J, Golab F, Morvaridzadeh M, Potter E, Akbari-Fakhrabadi M, Farsi F et al (2020)
The effects of curcumin supplementation on oxidative stress, Sirtuin-1 and peroxisome pro-
liferator activated receptor γ coactivator 1α gene expression in polycystic ovarian syndrome
(PCOS) patients: a randomized placebo-controlled clinical trial. Diabetes Metab Syndr
14(2):77–82. https://doi.org/10.1016/j.dsx.2020.01.002
26. Jamilian M, Foroozanfard F, Kavossian E, Aghadavod E, Shafabakhsh R, Hoseini A et al
(2020) Effects of curcumin on body weight, glycemic control and serum lipids in women
with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled trial. Clin
Nutr ESPEN 36:128–133. https://doi.org/10.1016/j.clnesp.2020.01.005
27. Sohaei S, Amani R, Tarrahi MJ, Ghasemi-Tehrani H (2019) The effects of curcumin supple-
mentation on glycemic status, lipid profile and hs-CRP levels in overweight/obese women
with polycystic ovary syndrome: a randomized, double-blind, placebo-controlled clinical
trial. Complement Ther Med 47:102201. https://doi.org/10.1016/j.ctim.2019.102201
28. Kia SJ, Basirat M, Saedi HS, Arab SA (2021) Effects of nanomicelle curcumin capsules on
prevention and treatment of oral mucositis in patients under chemotherapy with or with-
out head and neck radiotherapy: a randomized clinical trial. BMC Complement Med Ther
21(1):232. https://doi.org/10.1186/s12906-021-03400-4
29. Rodrigues HCN, Martins TFP, Santana N, Braga CC, Silva MAC, Cunha LCD et al (2021)
Antioxidant and anti-inflammatory response to curcumin supplementation in hemodi-
alysis patients: a randomized, double-blind, placebo-controlled clinical trial. Clin Nutr
ESPEN. 44:136–142. https://doi.org/10.1016/j.clnesp.2021.06.006
30. Vafadar-Afshar G, Rasmi Y, Yaghmaei P, Khadem-Ansari MH, Makhdoomi K, Rasouli
J (2021) The effects of nanocurcumin supplementation on inflammation in hemodialysis
patients: a randomized controlled trial. Hemodial Int 25(2):232–239. https://doi.org/10.1111/
hdi.12911
31. Salarolli RT, Alvarenga L, Cardozo L, Teixeira KTR, de SGML LJD et al (2021) Can curcumin
supplementation reduce plasma levels of gut-derived uremic toxins in hemodialysis patients?
A pilot randomized, double-blind, controlled study. Int Urol Nephrol 53(6):1231–1238.
https://doi.org/10.1007/s11255-020-02760-z
1110 M. Noormohammadi and F. Shidfar
metabolic syndrome and related disorders: a systematic review and meta-analysis of random-
ized controlled trials. Curr Pharm Des 24(27):3184–3199
49. He H-J, Wang G-Y, Gao Y, Ling W-H, Yu Z-W, Jin T-R (2012) Curcumin attenuates Nrf2
signaling defect, oxidative stress in muscle and glucose intolerance in high fat diet-fed mice.
World J Diabetes 3(5):94
50. Anh NH, Kim SJ, Long NP, Min JE, Yoon YC, Lee EG et al (2020) Ginger on human health:
a comprehensive systematic review of 109 randomized controlled trials. Nutrients 12(1):157
51. Khandouzi N, Shidfar F, Rajab A, Rahideh T, Hosseini P, Mir TM (2015) The effects of gin-
ger on fasting blood sugar, hemoglobin a1c, apolipoprotein B, apolipoprotein a-I and malo-
ndialdehyde in type 2 diabetic patients. Iran J Pharm Res 14(1):131–140
52. Shidfar F, Rajab A, Rahideh T, Khandouzi N, Hosseini S, Shidfar S (2015) The effect of gin-
ger (Zingiber officinale) on glycemic markers in patients with type 2 diabetes. J Complement
Integr Med 12(2):165–170. https://doi.org/10.1515/jcim-2014-0021
53. Carvalho GCN, Lira-Neto JCG, Araújo MFM, Freitas R, Zanetti ML, Damasceno
MMC (2020) Effectiveness of ginger in reducing metabolic levels in people with dia-
betes: a randomized clinical trial. Rev Lat Am Enfermagem 28:e3369. https://doi.
org/10.1590/1518-8345.3870.3369
54. Aryaeian N, Shahram F, Mahmoudi M, Tavakoli H, Yousefi B, Arablou T et al (2019) The
effect of ginger supplementation on some immunity and inflammation intermediate genes
expression in patients with active Rheumatoid Arthritis. Gene 698:179–185. https://doi.
org/10.1016/j.gene.2019.01.048
55. Mahluji S, Ostadrahimi A, Mobasseri M, Attari VE, Payahoo L (2013) Anti-inflammatory
effects of Zingiber officinale in type 2 diabetic patients. Adv Pharm Bull 3(2):273
56. Madkor HR, Mansour SW, Ramadan G (2011) Modulatory effects of garlic, ginger, turmeric
and their mixture on hyperglycaemia, dyslipidaemia and oxidative stress in streptozotocin-
nicotinamide diabetic rats. Br J Nutr 105(8):1210–1217. https://doi.org/10.1017/
s0007114510004927
57. Abdulrazaq NB, Cho MM, Win NN, Zaman R, Rahman MT (2012) Beneficial effects of gin-
ger (Zingiber officinale) on carbohydrate metabolism in streptozotocin-induced diabetic rats.
Br J Nutr 108(7):1194–1201. https://doi.org/10.1017/s0007114511006635
58. Shanmugam K, Ramakrishana C, Mallikarjuna K, Reddy KS (2009) The impact of ginger
on kidney carbohydrate metabolic profiles in STZ induced diabetic rats. Asian J Exp Sci
23(1):127–134
59. Rani MP, Padmakumari KP, Sankarikutty B, Cherian OL, Nisha VM, Raghu KG (2011)
Inhibitory potential of ginger extracts against enzymes linked to type 2 diabetes, inflamma-
tion and induced oxidative stress. Int J Food Sci Nutr 62(2):106–110. https://doi.org/10.310
9/09637486.2010.515565
60. Li Y, Tran VH, Duke CC, Roufogalis BD (2012) Gingerols of Zingiber officinale enhance
glucose uptake by increasing cell surface GLUT4 in cultured L6 myotubes. Planta Med
78(14):1549–1555. https://doi.org/10.1055/s-0032-1315041
61. Zhang XF, Tan BK (2000) Effects of an ethanolic extract of Gynura procumbens on serum
glucose, cholesterol and triglyceride levels in normal and streptozotocin-induced diabetic
rats. Singap Med J 41(1):9–13
62. Mohit M, Nouri M, Samadi M, Nouri Y, Heidarzadeh-Esfahani N, Venkatakrishnan K et al
(2021) The effect of sumac (Rhus coriaria L.) supplementation on glycemic indices: a system-
atic review and meta-analysis of controlled clinical trials. Complement Ther Med 61:102766
63. Shidfar F, Rahideh ST, Rajab A, Khandozi N, Hosseini S, Shidfar S et al (2014) The effect of
sumac (Rhus coriaria L.)powder on serum glycemic status, ApoB, ApoA-I and total antioxi-
dant capacity in type 2 diabetic patients. Iran. J Pharm Res 13(4):1249–1255
64. Rahideh ST, Shidfar F, Khandozi N, Rajab A, Hosseini SP, Mirtaher SM (2014) The effect
of sumac (Rhus coriaria L.) powder on insulin resistance, malondialdehyde, high sensitive
C-reactive protein and paraoxonase 1 activity in type 2 diabetic patients. J Res Med Sci
19(10):933–938
1112 M. Noormohammadi and F. Shidfar
65. Kazemi S, Shidfar F, Ehsani S, Adibi P, Janani L, Eslami O (2020) The effects of sumac
(Rhus coriaria L.) powder supplementation in patients with non-alcoholic fatty liver dis-
ease: a randomized controlled trial. Complement Ther Clin Pract 41:101259. https://doi.
org/10.1016/j.ctcp.2020.101259
66. Hariri N, Darafshi Ghahroudi S, Jahangiri S, Borumandnia N, Narmaki E, Saidpour A (2020)
The beneficial effects of sumac (Rhus coriaria L.) supplementation along with restricted
calorie diet on anthropometric indices, oxidative stress, and inflammation in overweight or
obese women with depression: a randomized clinical trial. Phytother Res 34(11):3041–3051.
https://doi.org/10.1002/ptr.6737
67. Asgary S, Salehizadeh L, Keshvari M, Taheri M, Spence ND, Farvid MS et al (2018) Potential
cardioprotective effects of sumac capsule in patients with hyperlipidemia: a triple-blind ran-
domized, placebo-controlled crossover trial. J Am Coll Nutr 37(4):286–292. https://doi.org/1
0.1080/07315724.2017.1394237
68. Hajmohammadi Z, Heydari M, Nimrouzi M, Faridi P, Zibaeenezhad MJ, Omrani GR et al
(2018) Rhus coriaria L. increases serum apolipoprotein-A1 and high-density lipoprotein
cholesterol levels: a double-blind placebo-controlled randomized clinical trial. J Integr Med
16(1):45–50. https://doi.org/10.1016/j.joim.2017.12.007
69. Mohammadi S, Kouhsari SM, Feshani AM (2010) Antidiabetic properties of the ethanolic
extract of Rhus coriaria fruits in rats. Daru 18(4):270
70. Giancarlo S, Rosa LM, Nadjafi F, Francesco M (2006) Hypoglycaemic activity of two spices
extracts: Rhus coriaria L. and Bunium persicum Boiss. Nat Prod Res 20(9):882–886
71. Shabana MM, El Sayed AM, Yousif MF, El Sayed AM, Sleem AA (2011) Bioactive constitu-
ents from Harpephyllum caffrum Bernh. and Rhus coriaria L. Pharmacogn Mag 7(28):298
72. Sales PM, Souza PM, Simeoni LA, Magalhães PO, Silveira D (2012) α-Amylase inhibitors:
a review of raw material and isolated compounds from plant source. J Pharm Pharm Sci
15(1):141–183
73. IuN N, Avezov G (1992) The efficacy of quercetin in alloxan diabetes. Eksp Klin Farmakol
55(1):42–44
74. Song Y, Manson JE, Buring JE, Sesso HD, Liu S (2005) Associations of dietary flavonoids
with risk of type 2 diabetes, and markers of insulin resistance and systemic inflammation in
women: a prospective study and cross-sectional analysis. J Am Coll Nutr 24(5):376–384.
https://doi.org/10.1080/07315724.2005.10719488
75. Capcarova M, Slamecka J, Abbas K, Kolesarova A, Kalafova A, Valent M et al (2012) Effects
of dietary inclusion of Rhus coriaria on internal milieu of rabbits. J Anim Physiol Anim Nutr
96(3):459–465
76. Asadi A, Shidfar F, Safari M, Hosseini AF, Fallah Huseini H, Heidari I et al (2019) Efficacy of
Melissa officinalis L. (lemon balm) extract on glycemic control and cardiovascular risk fac-
tors in individuals with type 2 diabetes: a randomized, double-blind, clinical trial. Phytother
Res 33(3):651–659. https://doi.org/10.1002/ptr.6254
77. Asadi A, Shidfar F, Safari M, Malek M, Hosseini AF, Rezazadeh S et al (2018) Safety and
efficacy of Melissa officinalis (lemon balm) on ApoA-I, Apo B, lipid ratio and ICAM-1 in
type 2 diabetes patients: a randomized, double-blinded clinical trial. Complement Ther Med
40:83–88. https://doi.org/10.1016/j.ctim.2018.07.015
78. Javid AZ, Haybar H, Dehghan P, Haghighizadeh MH, Mohaghegh SM, Ravanbakhsh M
et al (2018) The effects of Melissa officinalis (lemon balm) in chronic stable angina on
serum biomarkers of oxidative stress, inflammation and lipid profile. Asia Pac J Clin Nutr
27(4):785–791. https://doi.org/10.6133/apjcn.022018.01
79. Shekarriz Z, Shorofi SA, Nabati M, Shabankhani B, Yousefi SS (2021) Effect of Melissa
officinalis on systolic and diastolic blood pressures in essential hypertension: a double-blind
crossover clinical trial. Phytother Res 35(12):6883–6892. https://doi.org/10.1002/ptr.7251
80. Nayebi N, Esteghamati A, Meysamie A, Khalili N, Kamalinejad M, Emtiazy M et al (2019)
The effects of a Melissa officinalis L. based product on metabolic parameters in patients with
Natural Compounds with Pharmacological Properties in Clinical Trials 1113
115. Qin B, Nagasaki M, Ren M, Bajotto G, Oshida Y, Sato Y (2004) Cinnamon extract prevents
the insulin resistance induced by a high-fructose diet. Horm Metab Res 36(02):119–125
116. Lee J-S, Jeon S-M, Park E-M, Huh T-L, Kwon O-S, Lee M-K et al (2003) Cinnamate sup-
plementation enhances hepatic lipid metabolism and antioxidant defense systems in high
cholesterol-fed rats. J Med Food 6(3):183–191
117. Achakzai JK, Anwar Panezai M, Kakar MA, Kakar AM, Kakar S, Khan J et al (2019) In
vitro anticancer MCF-7, anti-inflammatory, and Brine Shrimp Lethality Assay (BSLA) and
GC-MS analysis of Whole Plant Butanol Fraction of Rheum ribes (WBFRR). Biomed Res
Int 2019:3264846. https://doi.org/10.1155/2019/3264846
118. Ghafouri A, Hosseini S, Shidfar S, Kamalinejad M, AghaHosseini F, Heydari I et al (2020)
The effect of Aqueous, Ethanolic extracts of Rheum ribes on insulin sensitivity, inflamma-
tion, oxidative stress in patients with type 2 diabetes mellitus: a randomized, double-blind,
placebo-controlled trial. J Herbal Medicine 24:100389
119. Khiveh A, Hashempur MH, Shakiba M, Lotfi MH, Shakeri A, Kazemeini S et al (2017)
Effects of rhubarb (Rheum ribes L.) syrup on dysenteric diarrhea in children: a randomized,
double-blind, placebo-controlled trial. J Integr Med 15(5):365–372
120. Sayyah M, Boostani H, Pakseresht S, Malayeri A (2009) Efficacy of hydroalcoholic extract
of Rheum ribes L. in treatment of major depressive disorder. J Med Plant Res 3(8):573–575
121. Fallah Huseini H, Heshmat R, Mohseni F, Jamshidi AH, Alavi S, Ahvasi M et al (2008)
The efficacy of rheum ribes L. stalk extract on lipid profile in hypercholesterolemic type II
diabetic patients: a randomized, double-blind, placebo-controlled, clinical trial. J Med Plant
7(27):92–97
122. Naqishbandi AM, Josefsen K, Pedersen ME, Jäger AK (2009) Hypoglycemic activity of Iraqi
Rheum ribes root extract. Pharm Biol 47(5):380–383
123. Matsuda H, Tomohiro N, Hiraba K, Harima S, Ko S, Matsuo K et al (2001) Study on anti-
Oketsu activity of rhubarb II. Anti-allergic effects of stilbene components from Rhei undu-
lati Rhizoma (dried rhizome of Rheum undulatum cultivated in Korea). Biol Pharm Bull
24(3):264–267
124. Hu B, Zhang H, Meng X, Wang F, Wang P (2014) Aloe-emodin from rhubarb (Rheum rhabar-
barum) inhibits lipopolysaccharide-induced inflammatory responses in RAW264. 7 macro-
phages. J Ethnopharmacol 153(3):846–853
125. Ziaei R, Foshati S, Hadi A, Kermani MAH, Ghavami A, Clark CC et al (2020) The effect of
nettle (Urtica dioica) supplementation on the glycemic control of patients with type 2 diabe-
tes mellitus: a systematic review and meta-analysis. Phytother Res 34(2):282–294
126. Nematgorgani S, Agah S, Shidfar F, Janani L, Faghihi A, Hosseini S (2020) The effect of
Urtica Dioica leaf extract intake on serum TNF-α, stool calprotectin and erythrocyte sedimen-
tation rate in patients with inflammatory bowel disease: a double-blind, placebo-controlled,
randomized, clinical trial. Mediterr J Nutr Metab 13(1):75–87
127. Nematgorgani S, Agah S, Shidfar F, Gohari M, Faghihi A (2017) Effects of Urtica dioica leaf
extract on inflammation, oxidative stress, ESR, blood cell count and quality of life in patients
with inflammatory bowel disease. Journal of Herbal Medicine 9:32–41
128. Akbar Karami A, Sheikhsoleimani M, Reza Memarzadeh M, Haddadi E, Bakhshpour M,
Mohammadi N et al (2020) Urtica Dioica root extract on clinical and biochemical parameters
in patients with benign prostatic hyperplasia, randomized controlled trial. Pak J Biol Sci
23(10):1338–1344. https://doi.org/10.3923/pjbs.2020.1338.1344
129. Namazi N, Esfanjani AT, Heshmati J, Bahrami A (2011) The effect of hydro alcoholic Nettle
(Urtica dioica) extracts on insulin sensitivity and some inflammatory indicators in patients
with type 2 diabetes: a randomized double-blind control trial. Pak J Biol Sci 14(15):775–779.
https://doi.org/10.3923/pjbs.2011.775.779
130. Namazi N, Tarighat A, Bahrami A (2012) The effect of hydro alcoholic nettle (Urtica dioica)
extract on oxidative stress in patients with type 2 diabetes: a randomized double-blind clinical
trial. Pak J Biol Sci 15(2):98–102. https://doi.org/10.3923/pjbs.2012.98.102
1116 M. Noormohammadi and F. Shidfar
Abbreviations
1 Introduction
Medicinal plants have been making an important role in humans. They are a very
rich source of traditional medicines. Nowadays, these medicinal plants are abun-
dantly used for modern medicines. For various diseases, medicinal plants could
make their milestone in treatment. The secondary metabolites produced by the
medicinal plants are very surprising and it is responsible for various biological
activities. Awareness of medicinal plant usage is an upshot of the many years of
struggle against illnesses due to which man learned to follow drugs in barks, seeds,
fruit bodies and other plant parts [1].
Genus Terminalia belonging to the family Combretaceae is spread worldwide
with 250 species, mainly in South Asia, Australia and South Africa. Among these
about 50 species are used as food [2]. Several Terminalia species revealed
nutraceutical value with the number of healthy profits, including the treatment for
various ailments [3]. Fruits of T. bellirica (Gaertn.) Roxb. and T. chebula Retz. are
common in Triphala, which is the polyherbal preparation in Ayurvedic and Thai folk
medicine by its pharmacological applications [4]. Terminalia species exhibit world-
wide distribution. The ethnobotanical importance and its medicinal use are various.
The majority of the fruits, leaves and stem bark of different Terminalia spp. has
been used for the treatment of geriatric diseases, memory improvement, abdominal
and back pain, cough, and colds, conjunctivitis, diarrhoea, fever, headache, heart
disorders, inflammation, leprosy, pneumonia, sexually transmitted diseases, urinary
disorders, etc. [5–7].
Medicinal plants are rich sources of bioactive compounds. Terminalia species
exhibit various biological activities such as antibacterial, antifungal and antipara-
sitic. Additionally, the plant extract showed antidiabetic, anticancer and antioxidant
activity. Terminalia has high therapeutic potential and clinical approval owed by its
traditional applications. Various Terminalia species are used in the form of herbal
medicine to cure several diseases like headache, fever, pneumonia, flu, abdominal
and back pain, cough and cold, conjunctivitis, geriatric, cancer, to improve memory,
diarrhoea, heart disorder, leprosy, sexually transmitted diseases and urinary tract
disorders. Moreover, these plants possess significant bioactivities like antibacterial,
antifungal, anti-inflammatory, antiviral, antiretroviral, antioxidant and antiparasitic
activity [8].
The phytochemical study of Terminalia species revealed the presence of glyco-
sides, flavonoids, tannins, phenols, saponin, carbohydrates, proteins, etc. [9].
Moreover, pure compounds are also isolated from the Terminalia species [10].
Likewise, the bioactivity study on Terminalia species provides a surprising result in
the field of research. It has been found that the growth of airborne pathogens like
MDR Acinetobacter sp. and MDR Pseudomonas aeruginosa is reduced when they
were treated with Terminalia bellirica (Gaertn.) Roxb [11]. and Terminalia chebula
Retz [12]. fruit extracts. Also, some of the Terminalia species are used as an inevi-
table ingredient in traditional medicines for the treatment of Malaria [13]. The anti-
cancer activity of Terminalia species has been focused on cancer which is one of the
important human diseases caused by various activities. It is caused by uncontrolled
cell division and can easily spread from one organ to another. So, the anticancer
research on Terminalia species has become a milestone in anticancer therapy.
Furthermore, some chemotherapy agents that are presently used have plant origins.
So, the studies on searching new plant species that provide natural products having
potential anticancer activity are relevant. Current information obtained from the
Scopus and Web of Science databases (2010–2020) reveals the anticancer potential
of Terminalia species [8]. Reports revealed antiproliferative activity of aqueous
extracts of Terminalia chebula Retz. using human lung cancer, A and mouse lung
cancer LLC cell lines [14]. The extract can inhibit cell proliferation by inducing
apoptosis and cell cycle arrest by regulating the mitochondrial pathway. It was
mediated by the proteins of the Bcl-2 family and inducing the PARP cleavage as
well as promoting cytochrome c release into the cytoplasm. Similarly, the Terminalia
fruits have an inhibitory effect against Caco-2 cells (Human epithelial cell line).
Phytopharmacological Aspects of the Genus Terminalia 1119
However, only methanol and aqueous fruit extracts showed antiproliferative activity
on the HeLa cell line. Also, the ethanolic leaf extract of T. catappa L. from Taiwan
revealed antiproliferative effects on HeLa and SiHa cervical cancer cell lines [15].
The current investigation mainly aims to provide detailed information regarding the
phytopharmacological aspects of the taxa Terminalia.
2 Phytochemical Aspects
Phytochemicals are the major cause of biological activities. The bioactivities include
triterpenes, alkaloids and flavonoids [16]. Tannins and polyphenols are the other
active phytocompounds, which contribute to the bioactivity of Terminalia spp. [17].
Also, triterpenoids, flavonoids and aliphatic compounds have several bioactive
properties [18] (Fig. 1).
Terminalia chebula is commonly used for digestive problems and its fruits are
used for the treatment of acidity, heat burn, heart disease, constipation, ulcers, piles,
inflammation, dysentery and diarrhoea. Also, it can remove toxins from the body.
The major phytocomponents present in T. chebula are alkaloids, soluble phenols,
phlobatannins, reducing sugars, carbohydrates, glycosides, inulin, terpenoids, tan-
nins and saponins. But there is no flavonoid, naphthoquinone and starch [19].
T. chebula aqueous fruit extract has the immense availability of carbohydrates,
anthocyanins, phenols and oils [20] whereas the proteins were absent in it. The pres-
ence of steroids is determined by Prajapati et al. [21].
The presence of non-nutritive phytochemicals and the other secondary metabo-
lites may be the reason for the medicinal properties of the plant. The extracts of the
dried powder of the pericarp of the fruits of T. chebula in petroleum ether, chloro-
form, ethyl alcohol and water were tested for the presence of tannins, alkaloids,
steroids, triterpenoids, flavonoids, hydroxyanthraquinones, cardiac glycosides,
saponins, carbohydrates (glucose and fructose), proteins, amino acids, fixed oils and
fatty acids. Proteins, cardiac glycosides, fixed oils and fats were found to be absent
in all the extracts [22]. Tannin has a role in plant growth regulation. Similarly, flavo-
noids can provide health benefits through cell signalling pathways as well as anti-
oxidant potential.
Terminalia bellirica has surprising medicinal values and is used in Ayurveda to
eliminate all three doshas. The fruits are used for the treatment of anaemia, jaundice
and white leprosy whereas seeds are used to control vomiting and cure bronchitis.
Additionally, T. bellerica is used to destroy germs and eye diseases. The major phy-
tocomponents present in T. bellerica are alkaloids, soluble phenols, phlobatannins,
reducing sugars, carbohydrates, glycosides, inulin, terpenoids and tannins. But
saponins, flavonoids, naphthoquinone and starch were not found in T. bellerica [19].
1120 A. Pokkadath et al.
Phytochemistry
Terminalia paniculata
Pharmacology
Activities
Anti-oxidant activity
Anti-microbial activity
Anti-proliferative activity
Anti-inflammatory activity
The fruits of T. bellerica extracted with petroleum ether, chloroform, ethyl acetate
and 20% aqueous methanol (hydroalcohol) revealed several biologically active phy-
tocompounds such as alkaloids, tannins, flavonoids, carbohydrates, glycosides,
saponins, steroids, free amino acids, fats, fixed oils and starch [23]. Among these
extracts, ethyl acetate and hydroalcoholic extracts were significantly higher in com-
position than the others. This indicates that the majority of the phytochemicals are
polar. Additionally, the phytochemical evaluation of ethyl acetate and hydroalco-
holic extracts was almost similar.
The plant extracts of T. bellerica possess phytochemical activities which are sig-
nificant for the therapeutic index. The ethanolic leaf and fruit extract of T. bellerica
revealed the presence of alkaloids, phenolics, tannins, carbohydrates, glycosides,
steroids, saponin and proteins. Proteins are detected in both leaves and fruit extract,
but the presence of free amino acids was not detected. Phenolics are the most com-
mon phytocompounds found in T. bellerica leaves and fruit extract. The fruit
Phytopharmacological Aspects of the Genus Terminalia 1121
extracts exhibit the prominent amount of phenolic content than the leaves extract.
Also, the fruits of T. bellerica are the major source of flavonoids [24].
Terminalia arjuna is an important cardiotonic plant in Ayurveda. The cardiopro-
tective activity of T. arjuna bark is an important ayurvedic remedy described in
many ancient Indian medicinal literatures like Charaka Samhita and Astanga
Hridayam. It has many therapeutic values and is used since ancient times by tribal
people to cure diarrhoea, tubercular cough, asthma, earache, cleansing sores, dysen-
tery, ulcers, syphilitic infection, sex stimulation, skin disorders, relieving excessive
menstrual bleeding, leucorrhoea, angina and heart disease [25]. Preliminary phyto-
chemical analysis revealed the biologically active compounds present in high con-
centrations like phytosterols, lactones, flavonoids, phenolic compounds, tannins and
glycosides [26]. The bark extract has plenty of arjunic acid, arjunolic acid, arjun-
genin acid, arjunglycesides and terminic acid, glycosides, flavonoids, tannins,
oligomeric proanthocyanidins, minerals, etc. Preliminary phytochemical analysis of
T. arjuna bark with different solvents including cold acetone and methanol revealed
the presence of tannin, alkaloid, triterpenoid, flavonoid, phytosteroids and saponins
[27]. T. arjuna plant encompasses flavonoids and glycosides. Among these, flavo-
noids like arjunic acid, arjunolic acid, arjungenin acid and arjunglucosides are
strong antioxidants that play an important role in antihyperglycemic and analgesic
activity. Amino acid and resins are not found in the bark of T. arjuna. The phyto-
components like triterpenoids, flavonoids, tannins and phytosteroids hold substan-
tial antioxidant, antihyperglycemic and cardioprotective potential [28, 29].
Triterpenoids have the same upshot as vitamin C. It can reduce myocardial abnor-
malities, as well as pathological changes in biochemical markers caused by cyclo-
sporine A. Saponins responsible for the inotropic effects of T. arjuna [27]. Major
classes of compounds identified in Terminalia species are enlisted in Table 1.
Phytochemical analysis of Terminalia arjuna by GC–MS analysis exhib-
ited five peaks with the presence of 13 compounds. All the 13 chemical com-
pounds are biologically very active and include decamethylcyclopentasiloxane,
benzoic acid, 2,5-bis(trimethylsiloxy)-, trimethylsilyl ester, anethole, dodeca-
methylcyclohexasiloxane, 2H-1,4-benzodiazepin-2-one 7-chloro-1,3- dihydro-5-
phenyl-1-(trimethylsilyl), silane, dimethyl(dimethyl(dimethyl(2-isopropylphenoxy)
silyloxy)silyloxy)(2-isopropylphenoxy)-, tetradecamethylcycloheptasiloxane,
3-isopropoxy-1,1,1,7,7,7-hexamethyl-3,5,5-tris(trimethylsiloxy)tetrasiloxane,
3-butoxy-1,1,1,7,7,7-hexamethyl-3,5,5-tris(trimethylsiloxy)tetrasiloxane, 2,4-ditert-
butylphenol, globulol, cadinol, heptadecyl 3-chloropropanoate, palmitoleic acid,
methyl (9Z)-9-hexadecenoate, dipentyl phthalate, phthalic acid, butyl 8-chlorooctyl
ester, dibutyl phthalate, 8,14-seco-3,19-epoxyandrostane-8,14-dione and 17-ace-
toxy-3á-methoxy-4,4-dimethyl, 1-monolinoleoylglycerol trimethylsilyl ether,
8,14-Seco-3,19-epoxyandrostane-8,14-dione, 17-acetoxy-3á-methoxy-4,4-di-
methyl [30]. Moreover, GC–MS analysis of T. arjuna exhibits 25 compounds which
include phenolic derivatives, hydrocarbons, alcoholic compounds, flavonoids,
alkaloids, ketones, carbohydrates, fatty acid ester, alkene compounds and fatty
acids [31].
1122 A. Pokkadath et al.
β-Sitosterol [68]
Globulol [30]
Luteolin [34]
(continued)
1124 A. Pokkadath et al.
Table 2 (continued)
Plant name Phytoconstituent Structure
T. sericea Linoleic acid [69]
Anolignan B [70]
Lupeol [71]
Sericoside [72]
Friedelin [74]
But the hydrolysis product of chebulinic acid and the di-O-galloyl analogue was
found to be very less in T. chebula [35].
The promising availability of these phytoconstituents might be the reason for the
attractive pharmacological activities of the Terminalia species.
3 Pharmacological Activities
3.1 Antioxidant Activity
Biological systems always create destructive free radicals by various metabolic pro-
cesses. Antioxidant activity can suppress this issue by scavenging those free radi-
cals. Oxidation is an important process in living organisms for the necessity of
performing catabolism. Oxygen-centred free radicals and several reactive oxygen or
nitrogen species were formed inside the body by various activities and cause severe
diseases like atherosclerosis, diabetes, cancer, and cirrhosis [36]. Automobile
exhaust, radiation, air pollution, cigarette smoke, and pesticides are environmental
pollutants that may form the reasons to generate free radicals. Various reports
revealed that Terminalia species possess high antioxidant potential [37] (Fig. 2).
The fruit extract of T. paniculata has an appreciable ability to scavenge free radi-
cals. All the four different antioxidant assays such as DPPH, hydroxyl free radical
scavenging assay, nitric oxide free radical scavenging assay, and superoxide free
radical scavenging activity revealed a significant result on antioxidant efficiency.
All the assays possess dose-dependent activity. In DPPH assay, ascorbic acid was
used as the standard, which exhibit 20.399 μg/mL of IC50 value while the IC50
value of T. paniculata was 23.068 μg/mL which was almost similar to that of ascor-
bic acid. So, it means T. paniculata exhibits a significant antioxidant potential [38].
T. arjuna is a major source of natural antioxidants and it has been widely used in
ayurvedic and Yunani Systems of medicine [39, 40]. Reports revealed that the anti-
oxidant activity of T. arjuna extracts varied substantially and it depends on the
nature of solvents as well as raw materials for the extraction process. Methanolic
extract of plant parts was considered as the best one to exploit the potential antioxi-
dant components. Also, the leaves extract of T. arjuna was found to prevent oxida-
tion. So, this means T. arjuna is a potential source of antioxidant agents and that is
why has been used in food and pharmaceutical industries [41].
1126 A. Pokkadath et al.
Photosynthesis Stress
- .
O2 NO
.
OH .
NO2
ROS/RNS
H2O2
Instability in cell system
Antioxidants
Phenolic compounds are the good scavengers of free radicals. It can deactivate
free radicals such as DPPH radicals. It is a stable free radical with maximum absorp-
tion at 517 nm (emax = 9660/M/cm). The reactions with DPPH and extract should
neutralize the free radicals which have been accredited by the presence of phenolic
antioxidants. Also, in T. chebula the presence of compounds such as ascorbic acid,
gallic acid and ellagic acid showed a significant reactivity with DPPH radicals.
Therefore, it has been clearly understood that phenolic constituents of the extract
are significantly responsible to react with DPPH free radicals. The extract of T. che-
bula is an excellent antioxidant agent to scavenge DPPH radicals so that it may
avoid the cellular organelle damage from radiations. Consequently, earlier reports
reveal its use as a radioprotector also [42].
3.2 Antimicrobial Activity
3.3 Antiproliferative Activity
Cancer is a large group of diseases that can start in any organ if the cells grow
uncontrollably. According to World Health Organization, it is a non-communicable
disease and the second leading cause of death and causes a big economic problem
for patients all over the world. So, the study about antiproliferative activity on plants
will provide relief to the medical field. Genus Terminalia exhibits high amount of
antioxidant agents which in turn are responsible for the therapeutic effects.
Furthermore, some of the plants of the genus Terminalia have been reported to
exhibit anticancer activities. Triphala is an ayurvedic medicine, which is the combi-
nation of Terminalia bellirica (Gawrtn.) Roxb. and Terminalia chebula (Retz.)
Lyons, together with Phyllanthus emblica. It has cytotoxic potential against thymic
lymphoma cells, human breast cancer cell lines, human prostate cancer cell lines
and human pancreatic cancer cell lines [47].
Antiproliferative activity of the aqueous extract of T. chebula was performed on
human lung cancer A and mouse lung cancer LLC cell lines. It was found to inhibit
1128 A. Pokkadath et al.
cell proliferation and promoted anticancer activity. The cell proliferation was
induced by the apoptosis and cell cycle arrest by regulating the mitochondrial path-
way mediated by proteins of the Bcl-2 family and inducing the PARP cleavage and
encouraging the cytochrome c release into the cytoplasm [14]. Similarly, the fruit
extracts of T. ferdinandiana are very effective on the Caco-2 cells. However, metha-
nolic and aqueous fruit extracts of T. ferdinandiana revealed antiproliferative activ-
ity on HeLa cells. The ethanolic leaf extract of T. catappa exhibits good
antiproliferative activity in HeLa and SiHa cervical cancer cell lines [15].
An isolated hydrolysable tannin compound casuarinin obtained from the bark of
T. arjuna proved antiproliferative activity in human breast cancer cell lines (MCF-7
cells). Casuarinin can inhibit the growth of MCF-7 by blocking cell cycle progres-
sion in the G0/G1 phase and promoting apoptosis. P53 gene makes a significant role
in apoptosis and cell cycle by DNA damage [48]. The activities of various cyclin-
dependent kinases are inhibited by P21/WAFI. Also, it blocks the phosphorylation
of retinoblastoma (RB) protein and inhibits the G1-S phase transition [49]. When
the MCF-7 cells were treated with casuarinin, it had no effective changes in the
expression of P53 whereas the P21/WAFI level was improved by casuarinin treat-
ment. Moreover, the flow cytometric analysis revealed that casuarinin causes arrest
of MCF-7 cells in the G0/G1 phase. So, the inhibition of cell cycle progression will
increase the activity of P21/ WAFI protein expression. Therefore, the quantity of
P53 protein, as well as its phosphorylation state, can regulate the transcriptional
activity and play a crucial role in P53-mediated functions. Additionally, when the
MCF-7 cells were treated with caspase-8-inhibitor, the cell growth inhibition as
well as the apoptotic induction of casuarinin was found to be reduced. Thus, casua-
rinin is a promising chemopreventive agent to cure breast cancer [32].
T. bellerica has significant medicinal activity and hence it was used in Indian
traditional medicinal systems. The majority of it proves its antiproliferative activity.
T. bellerica extract is used to check the antiproliferative activity against oral squa-
mous cell carcinoma (OSCC). It is a head and neck cancer induced by the impact of
carcinogens as well as the epidemiological influences like tobacco chewing and
betel quid, diet and immunodeficiency caused by viral infections. It was revealed
that gallic acid in T. bellerica is an antioxidant agent which in turn scavenge the free
radicals. So, they inferred that the strong antioxidant potential of gallic acid in
T. bellerica extract could exhibit a significant antiproliferative activity. Also, the
potential activity of gallic acid in T. bellerica inhibits autophagy, which in turn
diminishes the fusion of autophagosome and lysosomes in OSCC. The inhibition of
autophagy improves the apoptosis in OSCC by T. bellerica extract. Furthermore,
T. bellerica fruit extract possesses antiproliferative activity on Shionogi 115, breast
cancer MCF–7, prostate cancer PC–3, DU–145, leukaemia HK–63, HOS–1, HSC–2
and HL–60 cells. Also, the T. bellerica extract could make effective inhibition on
Hep2, Cal33 and FaDu cells that depends on both time and concentration. Apoptotic
protein analysis by Western blot technique is explicated by the effects of T. bellerica
extract on Cal33 cells. Mitochondria help for the process of initiation and progres-
sion of apoptosis, by the expression of pro-apoptotic protein Bax and consecutive
Phytopharmacological Aspects of the Genus Terminalia 1129
3.4 Anti-inflammatory Activity
Many human diseases are caused by inflammation. New reports revealed that non-
communicable diseases share common pathophysiological mechanisms, as well as
oxidative stress and inflammation, which makes the onset and development of these
diseases [54]. Terminalia species with reported anti-inflammatory activity in vitro
may be due to the presence of phytochemicals. There is an effective outcome
between the anti-inflammatory and the structural properties of phytochemicals that
have already been reported [55, 56].
The anti-inflammatory activity of T. coriacea was experimented in albino Wistar
rats using both acute and chronic models, carrageenan-induced paw oedema and
cotton pellet-induced granuloma, respectively. T. coriacea extract was examined
with different concentrations (125, 250 and 500 mg/kg) by oral uptake of the leaf
extract. All the concentrations showed an excellent anti-inflammatory activity in a
dose-dependent manner. Similarly, the anti-inflammatory studies using the ethano-
lic fruit extracts were performed at different concentrations like 50 to 500 mg/kg on
1130 A. Pokkadath et al.
4 Conclusion
Reports revealed that the genus Terminalia possess plenty of phytochemicals with
tremendous biological activities. The major phytochemicals found in Terminalia
spp. are alkaloids, phenols, flavonoids, tannins, terpenoids, carbohydrates, glyco-
sides, phlobatannins etc. All these phytochemicals have various biological proper-
ties in several fields. Most of them have high therapeutic values. Some of them serve
as antioxidant agents. In conclusion, it can be inferred that Terminalia spp. exhibit
significant antimicrobial, anticancer and anti-inflammatory properties. Thus, the
genus Terminalia seems to possess an irreplaceable group of plant species that may
be used in the therapeutic field for future studies and medical treatment.
Phytopharmacological Aspects of the Genus Terminalia 1131
Acknowledgments The first author is grateful to University of Calicut (U. O. No. 12753/2019/
Admn) for providing the financial assistance. Second author acknowledges the financial grant sup-
ported by the Council of Scientific and Industrial Research (CSIR) in the form of Senior Research
Fellowship (09/043(0186)/2017-EMR-1).
References
1. Petrovska BB (2012) Historical review of medicinal plants’ usage. Pharmacogn Rev 6:1–5
2. Fan C, Dong Y, Xie Y et al (2015) Shikonin reduces TGF-b1-induced collagen production and
contraction in hypertrophic scar-derived human skin fibroblasts. Int J Mol Med 36:985–991
3. Cock I (2015) The medicinal properties and phytochemistry of plants of the genus Terminalia
(Combretaceae). Inflammopharmacology 23:203–229
4. Intharuksa A, Ando H, Miyake K et al (2016) Molecular analysis of Terminalia spp. distrib-
uted in Thailand and authentication of crude drugs from Terminalia plants. Biol Pharm Bull
39:492–501
5. Maulik SK, Katiyar CK (2010) Terminalia arjuna in cardiovascular diseases: making the tran-
sition from traditional to modern medicine in India. Curr Pharm Biotechnol 11:855–860
6. Maulik SK, Talwar KK (2012) Therapeutic potential of Terminalia arjuna in cardiovascular
disorders. Am J Cardiovasc Drugs 12:157–163
7. Afshari AR, Sadeghnia HR, Mollazadeh H (2016) A review on potential mechanisms of
Terminalia chebula in Alzheimer’s disease. Adv Pharmacol Sci, pp 1–15
8. Das G, Kim DY, Fan C et al (2020) Plants of the genus Terminalia: an insight on its biological
potentials, pre-clinical and clinical studies. Front Pharmacol 11:1–30
9. Abraham A, Mathew L, Samuel S (2014) Pharmacognostic studies of the fruits of Terminalia
bellirica (Gaertn.) Roxb. J Pharmacognosy Phytochem 3:45–52
10. Wright MH, Sirdaarta J, White A et al (2016) Bacillus anthracis growth inhibitory properties
of Australian Terminalia spp.: putative identification of low polarity volatile components by
GC-MS headspace analysis. Pharm J 8:281–289
11. Dharmaratne MPJ, Manoraj A, Thevanesam V et al (2018) Terminalia bellirica fruit extracts:
in-vitro antibacterial activity against selected multidrug-resistant bacteria, radical scavenging
activity and cytotoxicity study on BHK-21 cells. BMC Complement Altern Med 18:325–325
12. Sharma C, Aneja KR, Kasera R et al (2012) Antimicrobial potential of Terminalia chebula
Retz. Fruit extracts against ear pathogens. WJOHNS 2:8–13
13. Malterud KEJP (2017) Ethnopharmacology, chemistry and biological properties of four
Malian medicinal plants. Plan Theory 6:2–13
14. Wang M, Yang L, Ji M et al (2015) Aqueous extract of Terminalia chebula induces apoptosis
in lung cancer cells via a mechanism involving mitochondria-mediated pathways. Braz Arch
Biol Technol 58:208–215
15. Lee CY, Yang SF, Wang PH et al (2019) Antimetastatic effects of Terminalia catappa leaf
extracts on cervical cancer through the inhibition of matrix metalloprotein-9 and MAPK path-
way. Environ Toxicol 34:60–66
16. Mongalo N, Mcgaw L, Segapelo T et al (2016) Ethnobotany, phytochemistry, toxicology and
pharmacological properties of Terminalia sericea Burch. Ex DC. (Combretaceae)–a review. J
Ethnopharmacol 194:789–802
17. Li K, Diao Y, Zhang H et al (2011) Tannin extracts from immature fruits of Terminalia che-
bula Fructus Retz. Promote cutaneous wound healing in rats. BMC Complement Altern Med
11:86–86
18. Chang Z, Zhang Q, Liang W et al (2019) A comprehensive review of the structure elucidation
of tannins from Terminalia Linn. Evidence-Based Complement Altern Med 2019:1–26
1132 A. Pokkadath et al.
41. Chatha SAS, Hussain AI, Asad R et al (2014) Bioactive components and antioxidant properties
of Terminalia arjuna L. extracts. J Food Process Technol 5:1–5
42. Naik GH, Priyadarsini KI, Naik DB et al (2004) Studies on the aqueous extract of Terminalia
chebula as a potent antioxidant and a probable radioprotector. Phytomedicine 11:530–538
43. Elizabeth KM (2005) Antimicrobial activity of Terminalia bellerica Indian J. Clin Biochem
20:150–153
44. Silva O, Duarte A, Pimentel M et al (1997) Antimicrobial activity of Terminalia macroptera
root. J Ethnopharmacol 57:203–207
45. Nair R, Chanda S (2008) Antimicrobial activity of Terminalia catappa, Manilkara zapota and
Piper betel leaf extract. Indian J Pharm Sci 70:390
46. Aneja KR, Sharma C, Joshi R (2012) Antimicrobial activity of Terminalia arjuna Wight &
Arn.: an ethnomedicinal plant against pathogens causing ear infection. Braz J Otorhinolaryngol
78:68–74
47. Gu B, Shalom J, Cock IE (2018) Anti-proliferative properties of Terminalia sericea burch. Ex
dc leaf extracts against Caco2 and HeLa cancer cell lines. Pharm J 10:408–415
48. May P, May E (1999) Twenty years of p53 research: structural and functional aspects of the
p53 protein. Oncogene 18:7621–7636
49. Harper JW, Adami GR, Wei N et al (1993) The p21 Cdk-interacting protein Cip1 is a potent
inhibitor of G1 cyclin-dependent kinases. Cell 75:805–816
50. Patra S, Panda PK, Naik PP et al (2020) Terminalia bellirica extract induces anticancer activity
through modulation of apoptosis and autophagy in oral squamous cell carcinoma. Food Chem
Toxicol 136:111073
51. Pinmai K, Chunlaratthanabhorn S, Ngamkitidechakul C (2008) Synergistic growth inhibitory
effects of Phyllanthus emblica and Terminalia bellerica extracts with conventional cytotoxic
agents: doxorubicin and cisplatin against human hepatocellular carcinoma and lung cancer
cells. World J Gastroenterol 14:1491–1497
52. Tan AC, Konczak I, Ramzan I (2011) Potential antioxidant, anti-inflammatory, and proapop-
totic anticancer activities of Kakadu plum and Illawarra plum polyphenolic fractions. Nutr
Cancer 63:1074–1084
53. Courtney R, Sirdaarta J, White A (2017) Inhibition of Caco-2 and HeLa proliferation by
Terminalia carpentariae C.T.White and Terminalia grandiflora Benth. Extracts: identification
of triterpenoid components. Pharm J 9:175–184
54. Camps J, Garcı́a-Heredia A (2014) Introduction: oxidation and inflammation, a molecular
link between non-communicable diseases. In: Oxidative Stress and Inflammation in non-
communicable diseases-molecular mechanisms and perspectives in therapeutics, Switzerland
55. Gautam R, Jachak SM (2009) Recent developments in antiinflammatory natural products. Med
Res Rev 29:767–820
56. Lago JHG, Toledo-Arruda AC, Mernak M et al (2014) Structure-activity association of flavo-
noids in lung diseases. Molecules 19:3570–3595
57. Khan MSA, Khatoon N, Al-Sanea MM et al (2018) Methanolic extract of leathery murdah,
Terminalia coriacea (Roxb.) Wight and Arn. Leaves exhibits antiinflammatory activity in
acute and chronic models. Med Principles Pract 27:267–271
58. Dawe A, Talom B, Kapche G et al (2017) Termiglaucescin, a new polyhydroxy triterpene
glucoside from Terminalia glaucescens with antioxidant and antiinflammatory potential.
ZNCBDA 72:203–208
59. Nair JJ, Aremu AO, Van Staden J (2012) Antiinflammatory effects of Terminalia phanero-
phlebia (Combretaceae) and identification of the active constituent principles. South Afr J Bot
81:79–80
60. Venkatalakshmi P, Brindha P, Vellingiri V (2015) In vitro antioxidant and anti-inflammatory
studies on bark, wood and fruits of Terminalia catappa L. Int J Phytomed 7:246–253
61. Dwevedi A, Dwivedi R, Sharma YK (2016) Exploration of phytochemicals found in Terminalia
sp. and their antiretroviral activities. Pharmacognosy Rev 10:73–83
1134 A. Pokkadath et al.
Abbreviations
1 Introduction
The term “epigenetics” proposed by Conrad Hal Waddington in the year 1942 is the
study of heritable changes during cell division which brings about the modifications
to the genome resulting in altered gene expression without changes to the underly-
ing DNA sequence [4, 11]. The diverse epigenetic modifications include self-rein-
forcing and distinctive mechanisms namely DNA methylation, histone modifications,
and RNA-mediated gene silencing, which may have everlasting effect on the expres-
sion of critical genes [7] Fig. 1. The epigenetic scaffold maintains the proper func-
tioning and survival of cells through certain epigenetic modulators that modify the
DNA as well as histone proteins within the nucleosome complex to render “ON” or
“OFF” signals to genes [11]. The most important epigenetic modulators and their
mode of action which can be exploited for the development of cancer therapeutics
are briefly discussed here.
Fig. 1 Cellular epigenetic mechanisms involved in the initiation and commencement of cancer.
The development and progression of deadly cancer involves deregulation of the cell cycle, meth-
ylation of target genes by DNA methyltransferases, histone modifications (acetylation, phosphory-
lation, methylation, and ubiquitination) by histone modifiers and transcriptional repression of
tumor suppressor genes via polycomb repressive complexes
increased the 5-methylated cytosine (5-mC) [24]. Aforementioned are only a few
natural compounds that have an exemplary role in mediating DNA methylation.
More details about the plant extracts and the other plant-derived compounds which
may play an inevitable role in cancer prevention targeting the methylation status or
DNMTs are enlisted in Tables 1 and 2.
Histone modifications are the major modifications which take place posttranslation-
ally causing inaccessibility of DNA to bind with their transcription factors leading
to transcriptional alterations [25]. Histone deacetylases (HDACs), responsible for
deacetylation, are a class of conserved enzymes which catalyze the removal of ace-
tyl groups from lysine side chain residues of histone tails and play an indispensable
role in the transcription of genes to regulate migration and cellular proliferation [11,
26]. Histone acetyltransferases (HATs) bring about the acetylation of histone tail in
which the positive charge over histone proteins is neutralized and, finally, RNA
polymerase gets recruited, enabling gene transcription. These two enzymes play a
leading role in the regulation of gene transcription, malignant transformation, and
progression in a widespread variety of tumors [27]. Class I and II HDACs are con-
sidered to be general oncoproteins that interact with substrates and regulate gene
expression to promote tumorigenesis and cancer development [14, 28]. The deacet-
ylation of HDAC3 and depletion of HDAC 10 affects the G2/M transitions through
the regulation of cyclin A [26].
Protein methyltransferases are also important for regulation of gene expression
epigenetically via methylation of target histones (H2AR3, H3R2, H3R8, and H4R3)
as well as nonhistones’ proteins and have been involved in playing crucial roles in
human diseases, including progression of cancer [29]. The predominant type of
epigenetic modification, methylation of lysine results in the repression or derepres-
sion of transcription depending on the position that is being methylated [30]. To be
concise, histone modifications are a segment of an intricate network and control
chromatin organization, transcription of genes, DNA repair, and replication [31].
The impairment in the regulation of these cellular processes and equilibrium loss
might lead to the development of diseases such as cancer [27].
It is the need of time to develop effective HDAC inhibitors (HDACi) from natural
sources that can limit tumor growth and restrict abnormal proliferation of cells
finally attenuating the tumor burden. There are some safe and efficacious plant-
derived compounds that impose epigenetic regulation in cancer cells. This is evident
when treatment with a polyphenol named resveratrol results in an augmentation in
the acetylation levels of histone H3 and H4 (H3K9ac, H3K14ac, H4K5ac, H4K12ac,
and H4K16ac) in renal carcinoma cells (RCC) [32]. A similar increase in acetyla-
tion at H3 and H4 histones with a higher HAT activity and lower HDAC activity was
observed in sulforaphane-treated A375 melanoma cells [33] and RKO colon cancer
cells [34]. Detailed information regarding the efficient natural plant extracts or
Table 1 Anticancer efficacy of various plant extracts against different cancer types
Cells used with IC50
1140
Name of the plant Family Parts used Extract Doses used Cancer type concentration Epigenetic modifications References
Acorus calamus (Sweet Acoraceae Plant Ethanol 0–1250 μg/ml Prostate LNCaP ↓ Proliferation [68]
flag) (923 μg/mL) ↑ Cleaved PARP
↓ VEGF mRNA
Adenosma bracteosum Plantaginaceae Aerial part Ethanol 0–100 μg/mL Lung NCI-H460 ↓ Proliferation [69]
(Bonati) Liver (4.57 ± 0.32 μg/mL) ↑ Caspase-3 activity
HepG2
(5.67 ± 0.09 μg/mL)
Aeluropus lagopoides Poaceace Leaves Hexane 0–100 μg/mL Liver HepG2 [70]
(Mangrove grass) Ethyl acetate (24.29 ± 0.85 μg/mL)
⦸ G0/G1 phase
HepG2
(11.22 ± 0.67 μg/mL)
Alcea rosea (Hollyhock) Malvaceae Seeds Ethyl acetate 0–100 μg/mL Colon HCT116 ↑ Cleaved PARP [71]
SW480 ↑ Bax expression
Balb/c mice Go/G1 phase arrest
↓ Cyclin D1, EZH2
↓ β-catenin, Ki-67
Allium sativum (Garlic) Amaryllidaceae Bulb – 0–50 μg/mL Kidney A498 S phase arrest [72]
Lung A549 ↑ p53, Bax, Cas3, Cas9
↓ Bcl-2
Alpinia nantoensis Zingiberaceae Rhizome Ethanol 0–200 μg/mL Breast MDA-MB-231 ↓ PI3K/AKT and Ras-ERK [73]
Leaves (28.13 ± 2.43 μg/mL) activation
MDA-MB-231 ↓ EGFR activation
(36.05 ± 3.74 μg/mL)
Alstonia scholaris Apocynaceae Leaves Alkaloid 0–25 g/ml Cervical Hela ↓ Cell viability [74]
(Blackboard tree) fraction (5.53 g/ml)
Andrographis Acanthaceae Leaves Methanol 0–300 μg Melanoma A375 and B16F10 ↓ Bcl-2, cyclin B1 [75]
nallamalayana ↑ BAD, BAX, Caspase-3
↑ p53, p21
Annona cherimola Annonaceae Leaves Ethanol 0–696 μg/mL Breast MDA-MB-231 ↑ DNA fragmentation [76]
(Cherimoya) 0–692 μg/mL Leukemia (390.2 μg/mL) ↑ p21 and Bax/Bcl-2 ratio [77]
Monomac-1 ↑ DNA fragmentation
P. Soni et al.
(18.67 μg/ml)
(continued)
1141
Table 1 (continued)
1142
MCF-7, SiHa
(140 ± 0.8, 94 ± 6.4,
121 ± 4.6 μg/ml)
Clinacanthus Nutans Acanthaceae Leaves and Hexane 0–200 μg/mL A549, CNE1, HepG2 ↑ Cells in sub-G1 phase [95]
(Snake Grass) stems ↑ Caspases 8, 9, and 3/7
P. Soni et al.
Cells used with IC50
Name of the plant Family Parts used Extract Doses used Cancer type concentration Epigenetic modifications References
Citrus limon (Lemon) Rutaceae Juice Aqueous – Breast MDA-MB-231 ↑ JAK1, JAK2, TYK2, IRF7, [96]
IRF3
↑ JAK/STAT pathway
Crataegus aronia Rosaceae Leaves Hydro- 0–300 μg/ml Lung A549 ↓ PARP-1, caspase-3, Bcl2 [97]
(Azarole) alcoholic ↑ Cells in sub G0 phase
Crotalaria verrucosa Papilionaceae Leaves Aqueous 0–100 μg/mL Cervical Hela ↑ Cell death [98]
(Blue rattlepod) Prostate (7.07 μg/mL) ↑ DNA damage
DU145 ↓ Cell invasion and migration
(6.30 μg/mL)
Cyclamen pseudibericum Primulaceae Tuber Petroleum 0–1000 μg/mL Lung A549 ↑ miRNA miR-200c [42]
ether and (41.64 ± 2.35 μg/mL) ↓ Epithelial-mesenchymal
ethanol transition
Curculigo orchioides Hypoxidaceae Rhizome Aqueous 0–300 μg/mL Liver HepG2 ↓ Bcl-2 expression [99]
(Kali musli) ethyl acetate Cervical (133.44 ± 1.1 μg/ml) ↑ Caspase-3 and Caspase-8
Breast HeLa
(136.50 ± 0.8 μg/ml)
MCF-7
(145.09 μg/ml)
Dendrobium venustum Orchidaceae Stem Methanol 0–100 μM Lung NCI-H460 ↓ Src-STAT3-c-Myc [100]
NCI-H292 pathways
Eclipta alba Asteraceae Plant Methanol 0–500 μg/ml Colon HCT-116 cells ↓ Colony formation and [101]
(False daisy) (179 ± 0.81 μg/ml) migration
Altered morphology
Elephantopus scaber Asteraceae Plant Ethanol 0–2000 μg/ml Breast T47D ↓ Cell viability [102]
(Elephant’s foot) (58.36 ± 2.38 μg/ml) Sub G1 phase arrest
Altered cell morphology
Fragaria vesca Rosaceae Leaves Ellagitanin- 0–10,000 μg/ HCC HepG2 G2/M phase arrest [103]
(Wild strawberry) enriched mL ↓ EDU incorporation
fraction ↓ FAS expression
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic...
(continued)
1143
Table 1 (continued)
1144
Lawsonia inermis Lythraceae Leaves Ethanol 0–700 μg/ml Lung A549 (490 μg/ml) ↑ Chromatin condensation [118]
(Henna) Colon DLD1 (480 μg/ml) ↑ DNA fragmentation
Liver HepG2 (610 μg/ml)
Leucas aspera Lamiaceae Plant Methanol 0–0.25 mg/mL Prostate PC3 cells Changed cell morphology [119]
(Thumbai) ↑ Antimigratory effect
Linum usitatissumum Linaceae Seeds – 0–100 μM Leukemia KG-1 (60 μM) ↑ DNA fragmentation [120]
(Flax) Monomac-1 (90 μM)
Litchi chinensis Sapindaceae Seeds Aqueous 0–400 μg/mL Breast BT474 and ↓ Proliferation & migration [121]
(Litchi) MDA-MB-231 cells
Mangifera indica Anacardiaceae Fruit peel Hydro- 0–600 μg/mL Colon HCT116 [122]
⦸ EMT
phosphorylation
↑ p53 expression
Marsypopetalum Annonaceae Leaves Methanol 0–1000 μg/ml Cervical HepG2 ↑ DNA fragmentation [123]
modestum Liver (358.68 μg/ml) Cell cycle arrest
HeLa ↑ Cells in the SubG1 phase
(445.91 μg/ml)
Matricaria chamomilla Asteraceae Leaves Aqueous 0–100 μg/mL Lung A549 S phase arrest [124]
(German Chamomile) (62.82 μg/mL) ↓ Bcl-2
↑ Bax, Caspase 3 and 7
(continued)
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic...
1145
Table 1 (continued)
1146
Myrmecodia pendans Rubiaceae Stem Methanol – Breast HCC-1954 (16 ppm) ↓ Cell migration [133]
(Ant plant) Cervical MCF7 cells (60 ppm)
Hela cells (13 ppm)
Tuber Methanol 0–100 ppm Colon Caco-2 (24 ppm) ↓ Proliferation [134]
HCT-116 (30 ppm) ↓ Colony formation
Myrmecodia tuberosa Rubiaceae Plant Ethanol 0–1000 μg/ml Oral KB cells ↑ Apoptosis [135]
(Ant plant) (215 μg/ml)
Nelumbo nucifera Nelumbonaceae Leaves Aqueous 0–4 mg/ml Breast MDA-MB-231 ↓ RhoA, Rac1, and Cdc42 [136]
(Indian lotus) 4 T-1
↓ PKCα activation
⦸ ERK/p38 signaling
P. Soni et al.
Cells used with IC50
Name of the plant Family Parts used Extract Doses used Cancer type concentration Epigenetic modifications References
Nigella sativa Ranunculaceae Seeds PBS 0–400 μg/ml Breast MCF-7 ↑ Caspase 3 [137]
(Black cumin) (8.05 ± 0.22 μg/ml) ↓ Survivin
Ocimum sanctum Lamiaceae Leaves Aqueous 0–50 μg/ml Leukemia K562 cells ↑ Cytotoxicity [138]
(Holy basil) (30 μg/ml)
Piper longum (Long Piperaceae Leaves Aqueous 0–80 μg/mL Cervical HeLa ↑ Cytotoxicity [139]
pepper) (5.27 μg/mL) ↑ DNA fragmentation
Plectranthus amboinicus Lamiaceae Leaves Aqueous 0–50 μg/mL Oral KB cells ↓ Cell viability [140]
(Mexican mint) Phyllanthaceae TiO2 ↑ p53 generation blocking
Phyllanthus niruri (Gale nanoparticles the DNA synthesis
of the wind)
Polygonum cuspidatum Polygonaceae Root Ethanol 0–100 μM Liver Hep3B and HepG2 ↓ STAT3 signaling [141]
(Japanese knotweed) (IC50 = 3.69 ± 0.51 ~ Cell-cycle arrest at S and
20.36 ± 2.90 μM) G2/M phases
Polygonatum cyrtonema Asparagaceae Rhizome – 0–1100 μg/mL Cervical HeLa cells ↑ Caspase-3 activity [142]
Hua ↓ CDK1 and Cyclin B1
↑ Bcl - 2, FasL, and
Caspases-8, 9, 10
Premna serratifolia Lamiaceae Roots Aqueous 0–1000 μg/mL Liver HepG2 ↑ Apoptosis [143]
(Sarunai) ↓ Cell migration
Primula vulgaris Primulaceae Flower DMSO 0–270 μg/mL Cervical HeLa cells Cell cycle arrest at S phase [53]
(Primrose) (182.4 μg/mL)
Putranjiva roxburghii Putranjivaceae Seeds Aqueous 0–10 mg/mL Pancreatic PANC-1 ↑ Cytotoxicity [144]
(Lucky bean) Breast (0.36 mg/mL) ↑ Cell death
Colon MDA-MB 231 ↑ DNA fragmentation
(0.26 mg/mL)
HCT-116
(0.54 mg/mL)
Rhus tripartita Anacardiaceae Leaves Acetone 0–64 μg/ml Leukemia THP-1 cells [57]
(63.12 ± 3.24 μg/ml) ↑ Apoptosis
⦸ Cell cycle
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic...
(continued)
1147
Table 1 (continued)
1148
Strobilanthes crispa Acanthaceae Stem Hexane 0–200 μg/ml Liver HepG-2 ↑ Cytotoxic [150]
⦸ EMT
↓ Tumor weight
⦸ ERK-Akt signaling
↓ Ki-67 expression
Urtica dioica Urticaceae Leaves Methanol 0–100 μg/mL Lung H1299 ↓ Proliferation [156]
(Common Nettle) (52.3 μg/mL) G2/M phase arrest
A549 ↑ Cleaved PARP
(47.4 μg/mL) ↑ Death receptor DR5
Viola odorata (Sweet Violaceae Aerial parts Hydro- 0–1000 μg/ml Breast MCF7-derived ↓ Migration, colony [157]
violet) alcoholic mammospheres formation
(600 μg/mL) ↑ Caspase 3/7 and 8
SKBR3-derived ↓ Decreased size and depth
mammospheres of generated tumor in CAM
(400 μg/mL) of chicken embryo
Viscum articulatum Santalaceae Frozen plant Aqueous 0–150 μg/mL Leukemia Jurkat E6.1 [158]
(Leafless mistletoe) tissue (2.4 μg/ml) ↓ Bcl-2,
⦸ G2/M phase
signaling
⦸ PI3K/AKT/mTOR
Zea mays Poaceae Corn silk Methanol 0–1000 μg/ml Breast MCF-7 ↓ Cell viability [166]
(Maize) ↑ Apoptosis
↑ p53, Bax, Caspase-3, and
Caspase-9
↓ Bcl-2 genes
P. Soni et al.
Cells used with IC50
Name of the plant Family Parts used Extract Doses used Cancer type concentration Epigenetic modifications References
Zingiber cassumunar Zingiberaceae Rhizome Chloroform 0–200 μg/ml Leukemia CEMss ↑ Cytotoxicity [167]
(Cassumunar ginger) Cervical (9.20 ± 0.02 μg/ml)
HeLa
(<15 μg/ml)
Zingiber officinale Zingiberaceae Rhizome Methanol 0–120 μM Cervical HeLa Cell cycle arrest at G0/G1 [168]
(Ginger) (29.19 μM) ↓ Cyclin A and Cyclin D1
Activate AMPK
⦸ PI3K/AKT
Ziziphus Jujube Rhamnaceae Dried fruit Powder 5% or 10% Colon C57BL/6 mice [169]
(Jujuba) w/w ↑ Bax
⦸ Tumor progression
Activation of NF-κB/IL-6/
JAK1/STAT3 signaling
Semi dried Aqueous (0–3 mg/ml) Cervical OV2008 ↓ Bcl-2 and ↑ Bax [170]
fruits Breast (1.2 ± 0.03 mg/ml) Bax/Bcl-2 ratio
MCF-7
(1.8 ± 0.08b mg/ml)
Abbreviations- ↑: Increases; ↓: Decreases; ⦸: Inhibit
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic...
1151
Table 2 Anticancer efficacy of plant-derived phytochemicals against different cancer types
1152
Name of the compound Modifications Cancer Cells and doses used Mode of action References
Curcumin DNA methylation Breast T47D and HCC-38 ↑ TET1 and DNMT3 [20]
[(1E,6E)-1,7-bis(4-hydroxy-3- (5 and 10 μM) Modulation of miR-29b
methoxyphenyl) Breast MCF-7 cells ↓ Proliferation [171]
hepta-1,6-diene-3,5-dione)] (20 μM) ↓ GSTP1 promoter methylation
Breast MDA-MB-361 ↓ Sp1 and DNMT1 [172]
BALB/c nu/nu mice ↑ DLC1 demethylation
↑ DLC1 expression
Histone modification Blood Myeloproliferative ↑ SOCS1 and SOCS3 levels [173]
neoplasm cells ↓ HDAC activity and HDAC8
(20 μM) expression
Schwannoma (arising from RT4 Schwannoma cells Activated PARP, Cas-3 and 9 [176]
peripheral nervous sheaths) (10 and 20 μM) ↓ miRNA 350, miRNA 17–2-3p, let
7e-3p, miRNA1224, miRNA
466b-1-3p, miRNA 18a-5p, and
miRNA 322-5p
↑ miRNA122-5p, miRNA 3473,
miRNA182, and miRNA344a-3p
Apoptosis via miRNA 344a-3p
P. Soni et al.
Name of the compound Modifications Cancer Cells and doses used Mode of action References
Epigallocatechin gallate DNA methylation Breast MCF-7 and MDA-MB-231 ↓ DNA methylation [23]
(EGCG) cells (20 μM) Reactivated expression of SCUBE2
[[(2R,3R)-5,7-dihydroxy-2- ↓ DNMT expression
(3,4,5-trihydroxyphenyl)-3,4- Esophageal ECa109 cells ↓ Cell viability [177]
dihydro-2H-chromen-3-yl] (0–200 mg/l) ↑ p16 mRNA and protein expression
3,4,5-trihydroxybenzoate] Induce apoptosis via demethylation
of p16
Histone modification Endothelial HMEC-1 and HUVECs ↑ H3K9/14 ac, H3ac [178]
(25–200 μM) ↑ H3K4me3 and H3K9me3
Affects expression of HDAC5 and 7,
p300, CREBP, LSD1 or KMT2A
Prostate LNCaP cells ↓ Acetylation of Androgen receptor [179]
(20 μM) (AR)
↓ Hormone responsiveness of AR
miRNA regulation Nasopharyngeal TW01 and TW06 cells Modulates miR-296 [180]
(0–80 μM)
↓ Cell migration and invasion
⦸ STAT3 signaling
Name of the compound Modifications Cancer Cells and doses used Mode of action References
Genistein DNA methylation Cervical HeLa ↓ Global DNA methylation levels [22]
[5,7-dihydroxy-3-(4- (50 μM) ↓ DNMT1, DNMT3B DNMT3A
hydroxyphenyl) chromen-4-one] ↓ TSGs promoter methylation
HCC HepG2 ↓ DNMT1 level [182]
(22 μM) ↑ Apoptosis
Prostate Norwegian patients ↑ NOTCH3and [183]
(30 mg) JAG1 mRNAs
↓ MYC and ↑ PTEN activity
Histone modification Cervical HeLa ↓ HDAC, HMT H3K9 Activity [22]
(50 μM) ↓ HDAC5, HDAC1, HDAC6
↑ SETD5, SETD7, SETD6
Prostate LNCaP and PC3 ↑ HAT activity [184]
(50 μM) ↑ acetylated histones 3, 4, 2H3K4,
3H3K4
miRNA regulation Lung H292 and A549 Regulates circ_0031250/miR-873-5p/ [185]
(80 μM) FOXM1 axis
Prostate LNCaP, 22RV1, and DU145 Demethylates promoter CpG sites [186]
cells closest to the miR-200c/miR-141 loci
(40 μM) ↑ miR-200c expression
Breast MDA-MB-435 and Hs578t ↓ miR-155 [187]
cells ↑ FOXO3, PTEN, casein kinase, and
(0–25 μM) p27
apoptosis
⦸ Cell viability and induces
P. Soni et al.
Name of the compound Modifications Cancer Cells and doses used Mode of action References
Resveratrol DNA methylation Lung A549 cells Demethylates the ZFP36 promoter [188]
[5-[(E)-2-(4-hydroxyphenyl) (0–100 μM) ↓ DNMT1 expression
ethenyl] benzene-1,3-diol] ↓ Cell proliferation
Breast MDA-MB-231 cells ↓ Promoter hypermethylation [189]
(100 μM) ↑ DNA hypomethylation
Hypomethylated 1459 and 1547
genes after 24 and 48 h
Histone modification Renal ACHN cells ↓ Cellular migration [32]
(0–62.5 μg/mL) ↑ acH3K9, acH3K14, cH4K12,
acH4K5, and acH4K16
Prostate DU145 and PC3M cells ↓ MTA2, MTA3, HDAC1, and [190]
(0–100 μM) HDAC2 levels
Deactivates MTA1/HDACs complex
miR-200c
⦸ Epithelial-mesenchymal transition
(continued)
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic...
1155
Table 2 (continued)
1156
Name of the compound Modifications Cancer Cells and doses used Mode of action References
Sulforaphane DNA methylation Skin Female SKH-1 hairless Altered expression of Pik3cd, Matk, [193]
[1-isothiocyanato-4- mice (2 μmol) and Adm2
methylsulfinylbutane] Changes in promoter CpG
methylation status
↓ Tumor incidence and tumor number
Colon Caco-2 ↓ DNMT1 protein expression [194]
(10–30 μmol/l) ↓ Nrf2 promoter methylation
Activation of Nrf2
Cervical HeLa [195]
(2.5 μM) ↓ DNMT3B expression
⦸ DNMT activity
phytochemicals and their epigenetic mode of action in cancer cells are summarized
in Tables 1 and 2.
Noncoding RNAs (ncRNAs) are a type of any functional and dynamic RNA mole-
cules that are not translated into protein. It is predicted that above 60% of the total
genome is transcribed into ncRNAs [35]. Based on their sizes, it can be divided into
several types of RNAs such as snoRNAs, microRNAs, piRNAs, exRNAs, scaRNAs,
endogenous siRNAs, snRNAs, and long noncoding RNAs which are involved in
various cellular processes [36, 37]. MicroRNAs (miRNAs) are a class of small func-
tional molecules with an approximate 22 nucleotide sequence that play substantial
regulatory role in many cellular processes, including induction in pluripotency, cel-
lular apoptosis, cellular proliferation, and DNA damage [38]. miRNAs typically
bind with a partial complementary sequence located on the 3’ untranslated region
(3’-UTR) of the corresponding target mRNA that negatively regulates gene expres-
sion which brings about translational repression or degradation of target mRNA
leading to inhibition or activation of downstream signaling pathways [39].
Several medicinal plants are reported to exert their anticancerous action by regu-
lating a varied range of miRNAs [40]. One such mode of action was shown when
polysaccharides from the roots of Astragalus membranaceus inhibited the JNK
pathway and induced apoptosis via upregulation of miR-133a in human osteosar-
coma MG63 cells [41]. Another study revealed the anticancer potential of Cyclamen
pseudibericum tuber extracts as it hindered the migration and invasion of lung car-
cinoma A549 cells and resulted in an increase in the expression of miR-200c [42].
Many other medicinal plants and phytochemicals are well known to exert their
potent anticancer activity by reversing the epigenetic modifications or altering the
signaling pathways overall, leading to a favorable preventive role in cancer therapy
and are listed in Tables 1 and 2.
Polycomb group (PcG) proteins are a group of chromatin modulators that comprises
two disparate polycomb repressive complexes (PRCs), namely PRC1 and PRC2,
which bind and act in a sequential manner and affect the chromatin structure of criti-
cal target genes [43]. Initially, the catalytic subunit of PRC2 complex named
Enhancer of zeste homolog 2 (EZH2) trimethylates the lysine 27 residue on histone
H3 (H3K27me3) which along with the suppressor of zeste 12 homolog (SUZ12)
and Embryonic Ectoderm Development (EED) further anchors the PRC1 complex
to the chromatin. Afterwards, the catalytic subunit of the PRC1 complex named
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1159
RING1B ubiquitinates the lysine 119 residue on histone H2A (H2AK119Ub), mak-
ing a platform for its association with B cell-specific moloney murine leukemia
virus integration site 1 (BMI-1). This phenomenon eventually leads to the transcrip-
tional suppression of gene expression via the induction of chromatin compaction
[17, 43].
Several reports have shown the abnormal overexpression of EZH2 and BMI-1
often promotes the tumorigenesis of lung cancer, breast cancer, prostate, bladder,
oral squamous cell carcinoma, and colorectal cancer. EZH2 is associated with the
metastasis of lymph nodes and combative phenotype of breast cancers guided by the
epigenetic silencing complex PRC-2/EED-EZH2 [44].
With the alarming rise of varied cancer types, the scientific community is in an
urgent need to develop novel, cost-effective, and harmless naturally derived com-
pounds that will aid in the cancer therapy in affected individuals. Some reports show
the striking effects of potent plant extracts on PcG proteins, making them ideal
candidates to be used in cancer therapy. For instance, the use of stem extract from
Celastrus orbiculatus averted the proliferation and progression in human NPC cell
line 5-8F cells by inhibiting the EZH2 signaling pathway [45]. Exposure to a puri-
fied extract from white grape pomace exhibited its anticancer effect in a multidisci-
plinary manner. It reduced the expression levels of prosurvival factors, phosphorylated
Akt (p-Akt), and B-cell lymphoma 2 (Bcl-2) with an increment in the expression of
p73 in a dose-dependent manner in Jurkat cells. The extract also led to a noteworthy
reduction in the expression of PcG proteins EZH2 and BMI1 and decreased the
expression of HDACs that may further mediate its proapoptotic effect by skipping
the DNA repair machinery [46]. Many more studies which assert the mode of action
of diverse plant extracts in the prevention of cancer are mentioned in Tables 1 and 2.
In a rapidly dividing mammalian cells with a 24-h cycle, the G1 phase lasts just
about 11 h, the S phase lasts approximately 8 h, the G2 phase lasts nearly 4 h, and
the M phase lasts roughly 1 h. The cell cycle is regulated at three foremost cell cycle
checkpoints. The G1 checkpoint, also known as the restriction point, where the cells
are not allowed to progress into the S phase if the conditions are unfavorable. The
G2 checkpoint halts the entry of the cell into the mitotic phase (M-phase) if certain
conditions are not achieved as the G2 checkpoint ensures that all the genes have
been replicated and also the replicated DNA is not damaged. The M checkpoint,
also called the spindle checkpoint, is located near the end of the metaphase stage
which ensures all the sister chromatids are suitably attached to the spindle microtu-
bules [47]. In addition to the checkpoints, a close cooperation between cyclins,
cyclin-dependent kinases (CDKs), and cyclin-dependent kinase inhibitors (CKIs) is
essential to ensure the orderly progression of the cell to the next phase of the cell
cycle [48].
1160 P. Soni et al.
The diverse members of the cyclin and CDK families have their extended star-
ring role linked to epigenetic regulation. The EZH2 phosphorylation at threonine
350 (Thr350) mediated by CDK1 and CDK2 positively regulates the methyltrans-
ferase activity of EZH2 and enhances suppression of target loci resulting into the
increased cell proliferation [49]. The CDK1 and CDK2 activity is at its peak during
the S-M phase transition of cell cycle. Therefore, enhanced methyltransferase activ-
ity of EZH2 during this period ensures that after S-phase, the repressive mark
H3K27me3 is integrated into newly synthesized histones and is inherited by daugh-
ter cells during M-phase [50].
Nearly 50% of the drugs used in cancer prevention as part of chemotherapy are
of plant origin. The present scenario of cancer emergence at an alarming rate
requires an outstanding approach toward the chemoprevention including cell cycle
regulators since the tumor cells proliferate abnormally via derestricting the cell
cycle control over checkpoints. A few studies have proposed the cytotoxic behavior
of several plant extracts toward malignant cancer cells. The methanol extract of
Asparagus laricinus brought cell death in prostate cancer cell line, PC3 and breast
cancer cell line, MCF7 through apoptosis. Cell death was also observed via cell
cycle arrest at S and G2 phase in MCF7 cells and mitotic (G2/M) phase arrest in
PC3 cells when treated with dichloromethane extracts of Senecio asperulus [51]. In
HepG2 cells, exposure to an aqueous methanol fraction of Calystegia soldanella
resulted in G0/G1 phase arrest in a dose-dependent manner with a reduction in the
expression of cyclin E, cyclin D1, and cell cycle inhibitor p21. This fraction also
decreased the expression of retinoblastoma (RB) and its downstream regulator E2F
(transcription factor) [52]. The cytotoxic effect of flower extract from Primula vul-
garis was observed in HeLa cells via induction of cell cycle arrest at S phase in a
concentration-dependent pattern [53]. A wide range of plant extracts and phyto-
chemicals posing an imperative control over the cell cycle progression in cancerous
cells are described in Tables 1 and 2.
Certain lifestyle factors and environmental exposures are reported to play an impor-
tant role in the onset of cancer. A point to be considered here is that it is almost
impossible to control or quantify these factors in a real-life scenario or to model
them in a particular lab setup [62]. This is the reason behind the late diagnosis of the
disease due to the late-stage presentation and continues to hinder the possibilities of
an effective therapy. Identifying the respective oncogene and the probable signaling
pathways associated with it seems to be crucial for the success of cancer therapy [63].
The most evident and possible strategy for cancer management is clinical devel-
opment of specific epigenetic or signaling pathway inhibitors for instance, PI3K
inhibitors, Akt inhibitors, DNMT inhibitors, HDACs inhibitors, and miRNA inhibi-
tors from natural products. In this case, discerning and targeting the particular pro-
teins within the intricate intracellular signaling and developing their inhibitors will
be a risky and complex task. Another concerning fact regarding the concept of tar-
geting DNMTs is lack of specificity since the inhibiting agents may not be precise
for a particular hypermethylated gene and can finally hypomethylate the whole
genome. This can be resolved by the epigenomic profiling of cancer which will
reveal the role of epigenetic alterations in the commencement and progression of
tumorigenesis, ultimately leading to the early detection and therapeutic intervention
via pharmacological targeting using specific anticancer drugs or plant-derived natu-
ral compounds [64]. Combination approaches along with patient selection strategies
will assist in intensifying the practical effectiveness of inhibitors to understand and
overcome drug resistance of chemotherapy or chemo drugs used nowadays.
The term CSCs refers to the small subpopulations of tumorigenic cells that can
potentially initiate the development of tumor and may cause major reversions as
they are resistant to treatment. It is evident that they can be regenerated even under
the strong therapeutic pressure and reformed microenvironment [65]. Inference
from quite a few clinical data promises the selective targeting of CSCs as a probable
strategy for treating relapsed cancer with the aid of plant extracts and plant-derived
phytochemicals.
nutrient-responsive factors for growth as they impose the glycolytic assets leading
to oxidative metabolism and restrict the glutathione peroxidase activity (GPX) with
low glutathione levels [66].
It is proposed to follow a 4-day diet cycle in a weekly manner and consume a
vegan ketogenic diet (lower protein and carbohydrate content with higher lipid con-
tent) ahead of each round of chemo/radiation therapy. This could potentially reorga-
nize the systemic metabolism and provide an unfavorable environment for cancer
cells. This diet with the aforementioned composition and period of treatment might
vary the membrane chemistry of cells with high peroxidation index (PUFA enrich-
ment), lessen the sulfur-dependent antioxidant activity, and compel a metabolic
shift in cancer cells to mitochondrial metabolism [67]. This shows the imperative
role of plants and plant-based products in cancer prevention and maintenance.
4 Conclusion
Fig. 2 Schematic diagram representing the possible strategy that may aid in an effective can-
cer therapy
1164 P. Soni et al.
appropriate drug delivery system (DDS) that suitably administers the required med-
ication under the correct conditions and within the necessary timeframe, thus avoid-
ing any potential risks or adverse effects. Early detection of cancer involves both
screening and surveillance and serves to be a reliable approach to improve and
manage the fatal outcomes of the disease. The practice of using natural and plant-
based extracts or phytochemicals as epigenetic agents appears to be a more reassur-
ing system for cancer treatment in the upcoming era. Even though more
comprehensive in vitro and in vivo trials are mandatory to examine their efficacy
and mode of action for an advanced cancer therapy.
References
13. Søreide K (2017) Cancer epigenetics. In: Handbook of epigenetics. Elsevier, Amsterdam,
pp 519–534. https://doi.org/10.1016/b978-0-12-805388-1.00034-1
14. Soni P, Ghufran MS, Kanade SR (2018) Aflatoxin B1 induced multiple epigenetic modu-
lators in human epithelial cell lines. Toxicon 151:119–128. https://doi.org/10.1016/j.
toxicon.2018.07.011
15. Cao H, Wang L, Chen B et al (2016) DNA demethylation upregulated Nrf2 expression in
Alzheimer’s disease cellular model. Front Aging Neurosci 7:244. https://doi.org/10.3389/
fnagi.2015.00244
16. Zhang W, Xu J (2017) DNA methyltransferases and their roles in tumorigenesis. Biomark
Res 5:1. https://doi.org/10.1186/s40364-017-0081-z
17. Soni P, Ghufran MS, Olakkaran S et al (2021) Epigenetic alterations induced by aflatoxin B1:
an in vitro and in vivo approach with emphasis on enhancer of zeste homologue-2/p21 axis.
Sci Total Environ 762:143175. https://doi.org/10.1016/j.scitotenv.2020.143175
18. Fabianowska-Majewska K, Kaufman-Szymczyk A, Szymanska-Kolba A et al (2021)
Curcumin from turmeric rhizome: a potential modulator of DNA methylation machinery in
breast cancer inhibition. Nutrients 13(2):332. https://doi.org/10.3390/nu13020332
19. Zhang J, Yang C, Wu C et al (2020) DNA methyltransferases in cancer: biology, paradox,
aberrations, and targeted therapy. Cancers (Basel) 12(8):2123. https://doi.org/10.3390/
cancers12082123
20. Al-Yousef N, Shinwari Z, Al-Shahrani B et al (2020) Curcumin induces re-expression of
BRCA1 and suppression of γ synuclein by modulating DNA promoter methylation in breast
cancer cell lines. Oncol Rep 43(3):827–838. https://doi.org/10.3892/or.2020.7473
21. Sundaram MK, Ansari MZ, Al Mutery A et al (2018) Genistein induces alterations of epigen-
etic modulatory signatures in human cervical cancer cells. Anti Cancer Agents Med Chem
18(3):412–421. https://doi.org/10.2174/1871520617666170918142114
22. Sundaram MK, Unni S, Somvanshi P et al (2019) Genistein modulates signaling pathways
and targets several epigenetic markers in HeLa cells. Genes (Basel) 10(12):955. https://doi.
org/10.3390/genes10120955
23. Sheng J, Shi W, Guo H et al (2019) The inhibitory effect of (−)-epigallocatechin-3-gallate on
breast cancer progression via reducing SCUBE2 methylation and DNMT activity. Molecules
24(16):2899. https://doi.org/10.3390/molecules24162899
24. Abbaszadeh S, Rashidipour M, Khosravi P et al (2020) Biocompatibility, cytotoxicity, anti-
microbial and epigenetic effects of novel chitosan-based quercetin nanohydrogel in human
cancer cells. Int J Nanomed 15:5963–5975. https://doi.org/10.2147/IJN.S263013
25. Chappell G, Pogribny IP, Guyton KZ et al (2016) Epigenetic alterations induced by genotoxic
occupational and environmental human chemical carcinogens: a systematic literature review.
Mutat Res Rev Mutat Res 768:27–45. https://doi.org/10.1016/j.mrrev.2016.03.004
26. Li G, Tian Y, Zhu WG (2020) The roles of histone deacetylases and their inhibitors in cancer
therapy. Front Cell Dev Biol 8:576946. https://doi.org/10.3389/fcell.2020.576946
27. Pant K, Peixoto E, Richard S et al (2020) Role of histone deacetylases in carcinogenesis:
potential role in cholangiocarcinoma. Cell 9(3):780. https://doi.org/10.3390/cells9030780
28. Ceccacci E, Minucci S (2016) Inhibition of histone deacetylases in cancer therapy: lessons
from leukaemia. Br J Cancer 114:605–611. https://doi.org/10.1038/bjc.2016.36
29. Zhu F, Rui L (2019) PRMT5 in gene regulation and hematologic malignancies. Genes Dis
6(3):247–257. https://doi.org/10.1016/j.gendis.2019.06.002
30. Yao B, Christian KM, He C et al (2016) Epigenetic mechanisms in neurogenesis. Nat Rev
Neurosci 17(9):537–549. https://doi.org/10.1038/nrn.2016.70
31. Zhao S, Allis CD, Wang GG (2021) The language of chromatin modification in human can-
cers. Nat Rev Cancer 21:413–430. https://doi.org/10.1038/s41568-021-00357-x
32. Dai L, Chen L, Wang W et al (2020) Resveratrol inhibits ACHN cells via regulation of his-
tone acetylation. Pharm Biol 58(1):231–238. https://doi.org/10.1080/13880209.2020.173850
3
1166 P. Soni et al.
33. Mitsiogianni M, Trafalis DT, Franco R et al (2021) Sulforaphane and iberin are potent epi-
genetic modulators of histone acetylation and methylation in malignant melanoma. Eur J
Nutr 60:147–158. https://doi.org/10.1007/s00394-020-02227-y
34. Martin SL, Kala R, Tollefsbol TO (2018) Mechanisms for the inhibition of colon cancer
cells by sulforaphane through epigenetic modulation of microRNA-21 and human telomer-
ase reverse transcriptase (hTERT) down-regulation. Curr Cancer Drug Targets 18(1):97–106.
https://doi.org/10.2174/1568009617666170206104032
35. Zhang P, Wu W, Chen Q et al (2019) Non-coding RNAs and their integrated networks. J
Integr Bioinform 16(3):20190027. https://doi.org/10.1515/jib-2019-0027
36. Chen J, Xue Y (2016) Emerging roles of non-coding RNAs in epigenetic regulation. Sci
China Life Sci 59:227–235. https://doi.org/10.1007/s11427-016-5010-0
37. Zhu L, Zhang B, Dai Y et al (2017) A review: epigenetic mechanism in ochratoxin A toxicity
studies. Toxins (Basel) 9(4):113. https://doi.org/10.3390/toxins9040113
38. Mens MMJ, Ghanbari M (2018) Cell cycle regulation of stem cells by microRNAs. Stem Cell
Rev Rep 14:309–322. https://doi.org/10.1007/s12015-018-9808-y
39. O’Brien J, Hayder H, Zayed Y et al (2018) Overview of microRNA biogenesis, mechanisms
of actions, and circulation. Front Endocrinol (Lausanne) 9:402. https://doi.org/10.3389/
fendo.2018.00402
40. Shanmugapriya N, Sasidharan S (2020) MicroRNA deregulation and cancer and medici-
nal plants as microRNA regulator. Asian Pac J Trop Biomed 10:47–53. https://doi.
org/10.4103/2221-1691.275419
41. Chu Y, Fang Y, Chi J et al (2018) Astragalus polysaccharides decrease prolifera-
tion, migration, and invasion but increase apoptosis of human osteosarcoma cells by
up-regulation of microRNA-133a. Braz J Med Biol Res 51(12):e7665. https://doi.
org/10.1590/1414-431X20187665
42. Karagur ER, Ozay C, Mammadov R et al (2018) Anti-invasive effect of Cyclamen pseudi-
bericum extract on A549 non-small cell lung carcinoma cells via inhibition of ZEB1 medi-
ated by miR-200c. J Nat Med 72(3):686–693. https://doi.org/10.1007/s11418-018-1204-z
43. Yang Y, Li G (2020) Post-translational modifications of PRC2: signals directing its activity.
Epigenetics Chromatin 13:47. https://doi.org/10.1186/s13072-020-00369-1
44. Sun S, Yu F, Zhang L et al (2016) EZH2, an on-off valve in signal network of tumor cells. Cell
Signal 28(5):481–487. https://doi.org/10.1016/j.cellsig.2016.02.004
45. Wang X, Huang Y, Chen Y et al (2018) Efficacy of extracts of Celastrus orbiculatus in
suppressing migration and invasion by inhibiting the EZH2/ROCK1 signaling pathway in
human nasopharyngeal carcinoma. Oncol Lett 15(5):6695–6700. https://doi.org/10.3892/
ol.2018.8149
46. León-González AJ, Jara-Palacios MJ, Abbas M et al (2017) Role of epigenetic regulation on
the induction of apoptosis in Jurkat leukemia cells by white grape pomace rich in phenolic
compounds. Food Funct 8(11):4062–4069. https://doi.org/10.1039/c7fo00263g
47. Visconti R, Della Monica R, Grieco D (2016) Cell cycle checkpoint in cancer: a therapeuti-
cally targetable double-edged sword. J Exp Clin Cancer Res 35:153. https://doi.org/10.1186/
s13046-016-0433-9
48. Ding L, Cao J, Lin W et al (2020) The roles of cyclin-dependent kinases in cell-cycle progres-
sion and therapeutic strategies in human breast cancer. Int J Mol Sci 21(6):1960. https://doi.
org/10.3390/ijms21061960
49. Aleem E, Arceci RJ (2015) Targeting cell cycle regulators in hematologic malignancies.
Front Cell Dev Biol 3:16. https://doi.org/10.3389/fcell.2015.00016
50. Ma Y, Kanakousaki K, Buttitta L (2015) How the cell cycle impacts chromatin architecture
and influences cell fate. Front Genet 6:19. https://doi.org/10.3389/fgene.2015.00019
51. Mfengwana PH, Mashele SS, Manduna IT (2019) Cytotoxicity and cell cycle analysis of
Asparagus laricinus Burch. and Senecio asperulus DC. on breast and prostate cancer cell
lines. Heliyon 5(5):e01666. https://doi.org/10.1016/j.heliyon.2019.e01666
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1167
52. Lee JI, Kim IH, Nam TJ (2017) Crude extract and solvent fractions of Calystegia soldanella
induce G1 and S phase arrest of the cell cycle in HepG2 cells. Int J Oncol 50(2):414–420.
https://doi.org/10.3892/ijo.2017.3836
53. Demir S, Turan I, Aliyazicioglu R et al (2018) Primula vulgaris extract induces cell cycle
arrest and apoptosis in human cervix cancer cells. J Pharm Anal 8(5):307–311. https://doi.
org/10.1016/j.jpha.2018.05.003
54. Asati V, Mahapatra DK, Bharti SK (2016) PI3K/Akt/mTOR and Ras/Raf/MEK/ERK signal-
ing pathways inhibitors as anticancer agents: structural and pharmacological perspectives.
Eur J Med Chem 109:314–341. https://doi.org/10.1016/j.ejmech.2016.01.012
55. Manning BD, Toker A (2017) AKT/PKB signaling: navigating the network. Cell
169(3):381–405. https://doi.org/10.1016/j.cell.2017.04.001
56. Yang J, Nie J, Ma X et al (2019) Targeting PI3K in cancer: mechanisms and advances in clini-
cal trials. Mol Cancer 18:26. https://doi.org/10.1186/s12943-019-0954-x
57. Tlili H, Macovei A, Buonocore D et al (2021) The polyphenol/saponin-rich Rhus tripartita
extract has an apoptotic effect on THP-1 cells through the PI3K/AKT/mTOR signaling path-
way. BMC Complement Med Ther 21(1):153. https://doi.org/10.1186/s12906-021-03328-9
58. Nassan MA, Soliman MM, Ismail SA et al (2018) Effect of Taraxacum officinale
extract on PI3K/Akt pathway in DMBA-induced breast cancer in albino rats. Biosci Rep
38(6):BSR20180334. https://doi.org/10.1042/BSR20180334
59. Kim J, Jung KH, Ryu HW et al (2019) Apoptotic effects of Xanthium strumarium via PI3K/
AKT/mTOR pathway in hepatocellular carcinoma. Evid Based Complement Alternat Med
2019:2176701. https://doi.org/10.1155/2019/2176701
60. Pucci C, Martinelli C, Ciofani G (2019) Innovative approaches for cancer treatment: current
perspectives and new challenges. Ecancermedicalscience 13:961. https://doi.org/10.3332/
ecancer.2019.961
61. Malone ER, Oliva M, Sabatini PJB et al (2020) Molecular profiling for precision cancer
therapies. Genome Med 12(1):8. https://doi.org/10.1186/s13073-019-0703-1
62. Holleman GA, Hooge ITC, Kemner C et al (2020) The ‘real-world approach’ and its prob-
lems: a critique of the term ecological validity. Front Psychol 11:721. https://doi.org/10.3389/
fpsyg.2020.00721
63. Ankathil R (2019) The mechanisms and challenges of cancer chemotherapy resistance: a cur-
rent overview. Eur J Mol Clin Med 6(1):26–34. https://doi.org/10.5334/ejmcm.269
64. Lorscheider M, Gaudin A, Nakhlé J et al (2021) Challenges and opportunities in the deliv-
ery of cancer therapeutics: update on recent progress. Ther Deliv 12(1):55–76. https://doi.
org/10.4155/tde-2020-0079
65. Walcher L, Kistenmacher AK, Suo H et al (2020) Cancer stem cells-origins and biomark-
ers: perspectives for targeted personalized therapies. Front Immunol 11:1280. https://doi.
org/10.3389/fimmu.2020.01280
66. Salvadori G, Mirisola MG, Longo VD (2021) Intermittent and periodic fasting, hormones,
and cancer prevention. Cancers 13(18):4587. https://doi.org/10.3390/cancers13184587
67. Lettieri-Barbato D, Aquilano K (2018) Pushing the limits of cancer therapy: the nutrient
game. Front Oncol 8:148. https://doi.org/10.3389/fonc.2018.00148
68. Koca HB, Köken T, Özkurt M et al (2018) Effects of Acorus calamus plant extract on prostate
cancer cell culture. Anat J Bot 2(1):46–51. https://doi.org/10.30616/ajb.391985
69. Nguyen NH, Ta QTH, Pham QT et al (2020) Anticancer activity of novel plant extracts
and compounds from Adenosma bracteosum (Bonati) in human lung and liver cancer cells.
Molecules 25(12):2912. https://doi.org/10.3390/molecules25122912
70. Saleh KA, Albinhassan TH, Elbehairi SEI et al (2019) Cell cycle arrest in different cancer
cell lines (liver, breast, and colon) induces apoptosis under the influence of the chemical
content of Aeluropus lagopoides leaf extracts. Molecules 24(3):507. https://doi.org/10.3390/
molecules24030507
1168 P. Soni et al.
71. Ahmed I, Roy BC, Subramaniam D et al (2016) An ornamental plant targets epigenetic sig-
naling to block cancer stem cell-driven colon carcinogenesis. Carcinogenesis 37(4):385–396.
https://doi.org/10.1093/carcin/bgw009
72. Özkan İ, Koçak P, Yıldırım M et al (2021) Garlic (Allium sativum)-derived SEVs inhibit can-
cer cell proliferation and induce caspase-mediated apoptosis. Sci Rep 11:14773. https://doi.
org/10.1038/s41598-021-93876-4
73. Kuo CY, Weng TS, Kumar KJS et al (2019) Ethanol extracts of dietary herb, Alpinia
nantoensis, exhibit anticancer potential in human breast cancer cells. Integr Cancer Ther
18:1534735419866924. https://doi.org/10.1177/1534735419866924
74. Ahmad MS, Ahmad S, Ali A et al (2016) Anticarcinogenic and antimutagenic activity of
Alstonia scholaris on the albino mice bone marrow cells and peripheral human lympho-
cyte culture against methyl methane sulfonate induced genotoxicity. Adv Biomed Res 5:92.
https://doi.org/10.4103/2277-9175.183140
75. Purushotham G, Padma Y, Nabiha Y et al (2016) In vitro evaluation of anti-proliferative,
anti-inflammatory and pro-apoptotic activities of the methanolic extracts of Andrographis
nallamalayana Ellis on A375 and B16F10 melanoma cell lines. 3 Biotech 6(2):212. https://
doi.org/10.1007/s13205-016-0529-0
76. Younes M, Ammoury C, Haykal T et al (2020) The selective anti-proliferative and pro-apop-
totic effect of A. cherimola on MDA-MB-231 breast cancer cell line. BMC Complement Med
Ther 20(1):343. https://doi.org/10.1186/s12906-020-03120-1
77. Ammoury C, Younes M, El Khoury M et al (2019) The pro-apoptotic effect of a Terpene-rich
Annona cherimola leaf extract on leukemic cell lines. BMC Complement Altern Med 19:365.
https://doi.org/10.1186/s12906-019-2768-1
78. Köken T, Koca B, Özkurt M et al (2016) Apium graveolens extract inhibits cell proliferation
and expression of vascular endothelial growth factor and induces apoptosis in the human
prostatic carcinoma cell line LNCaP. J Med Food 19(12):1166–1171. https://doi.org/10.1089/
jmf.2016.0061
79. Ali S, Ejaz M, Dar KK et al (2020) Evaluation of chemopreventive and chemotherapeutic effect
of Artemisia vulgaris extract against diethylnitrosamine-induced hepatocellular carcinogen-
esis in Balb C mice. Braz J Biol 80(3):484–496. https://doi.org/10.1590/1519-6984.185979
80. Rajput S, Kumar D, Agrawal V (2020) Green synthesis of silver nanoparticles using Indian
Belladonna extract and their potential antioxidant, anti-inflammatory, anticancer and larvi-
cidal activities. Plant Cell Rep 39(7):921–939. https://doi.org/10.1007/s00299-020-02539-7
81. Mustafa K, Mohamed H, Shah AM et al (2020) In vitro anticancer potential of Berberis
lycium royle extracts against human hepatocarcinoma (HepG2) cells. Biomed Res Int
2020:8256809. https://doi.org/10.1155/2020/8256809
82. Dar KK, Ali S, Ejaz M et al (2019) In vivo induction of hepatocellular carcinoma by dieth-
ylnitrosoamine and pharmacological intervention in Balb C mice using Bergenia ciliata
extracts. Braz J Biol 79(4):629–638. https://doi.org/10.1590/1519-6984.186565
83. Chen X, Li S, Li D et al (2020) Ethanol extract of Brucea javanica seed inhibit triple-nega-
tive breast cancer by restraining autophagy via PI3K/Akt/mTOR pathway. Front Pharmacol
11:606. https://doi.org/10.3389/fphar.2020.00606
84. Naik Bukke A, Nazneen Hadi F, Babu KS et al (2018) In vitro studies data on anticancer
activity of Caesalpinia sappan L. heartwood and leaf extracts on MCF7 and A549 cell lines.
Data Brief 19:868–877. https://doi.org/10.1016/j.dib.2018.05.050
85. Sui M, Yang H, Guo M et al (2021) Cajanol sensitizes A2780/Taxol cells to paclitaxel by
inhibiting the PI3K/Akt/NF-κB signaling pathway. Front Pharmacol 12:783317. https://doi.
org/10.3389/fphar.2021.783317
86. Yumukjije C, Ndacyayisenga J (2019) Phytochemical analysis & in vitro anticancer activity
of methanol extract of Catharanthus Roseus leaves using HEPG2 cell line. IAETSD J Adv
Res Appl Sci vi(viii):2394–8442
87. Sudevan S, Paramasivam R, Sundar S et al (2017) Evaluation of metabolic compounds of
Catharanthus roseus and its anticancer activity. Eur J Pharm Med Res 4(9):282–290
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1169
88. Abouelela ME, Orabi MAA, Abdelhamid RA et al (2018) Chemical and cytotoxic investiga-
tion of non-polar extract from Ceiba Pentandra (L.) Gaertn.: a study supported by computer
based screening. J Appl Pharm Sci 8(07):057–064. https://doi.org/10.7324/JAPS.2018.8710
89. Hamid IS, Rai Widjaja NM, Damayanti R (2016) Anticancer activity of Centella asiatica
leaves extract in benzo(a)pyrene-induced mice. Int J Pharmacogn Phytochem Res 8(1):80–84.
ISSN: 0975-4873
90. Adedapo AA, Oyagbemi AA, Fagbohun OA et al (2016) Evaluation of the anticancer prop-
erties of the methanol leaf extract of Chromolaena odorata on HT29 lung cancer cell line.
FASEB J 30:1193.6-1193.6. https://doi.org/10.1096/fasebj.30.1_supplement.1193.6
91. Elkady AI (2019) Targeting prostate cancer cell proliferation, stemness and metastatic poten-
tial using Costus speciosus-derived phytochemicals. Am J Transl Res 11(4):2550–2569
92. Chen CY, Yen CY, Wang HR et al (2016) Tenuifolide B from Cinnamomum tenuifolium
stem selectively inhibits proliferation of oral cancer cells via apoptosis, ROS generation,
mitochondrial depolarization, and DNA damage. Toxins (Basel) 8(11):319. https://doi.
org/10.3390/toxins8110319
93. Perveen S, Ashfaq H, Ambreen S et al (2021) Methanolic extract of Citrullus colocynthis sup-
presses growth and proliferation of breast cancer cells through regulation of cell cycle. Saudi
J Biol Sci 28(1):879–886. https://doi.org/10.1016/j.sjbs.2020.11.029
94. Chowdhury K, Sharma A, Kumar S et al (2017) Colocynth extracts prevent epithelial to mes-
enchymal transition and stemness of breast cancer cells. Front Pharmacol 8:593. https://doi.
org/10.3389/fphar.2017.00593
95. Ng PY, Chye SM, Ng CHH et al (2017) Clinacanthus nutans hexane extracts induce apopto-
sis through a caspase-dependent pathway in human cancer cell lines. Asian Pac J Cancer Prev
18(4):917–926. https://doi.org/10.22034/APJCP.2017.18.4.917
96. Mahmood N, Younas H, Zafar M et al (2021) Effects of plants extracts on the expression of
major genes of JAK/STAT pathway. Nucleosides Nucleotides Nucleic Acids 40(4):434–469.
https://doi.org/10.1080/15257770.2021.1896000
97. Omairi I, Kobeissy F, Nasreddine S (2020) Anti-oxidant, anti-hemolytic effects of Crataegus
aronia leaves and its anti- proliferative effect enhance cisplatin cytotoxicity in A549 human
lung cancer cell line. Asian Pac J Cancer Prev 21(10):2993–3003. https://doi.org/10.31557/
APJCP.2020.21.10.2993
98. Sana SS, Kumbhakar DV, Pasha A et al (2020) Crotalaria verrucosa leaf extract-mediated
synthesis of zinc oxide nanoparticles: assessment of antimicrobial and anticancer activity.
Molecules 25(21):4896. https://doi.org/10.3390/molecules25214896
99. Hejazi II, Khanam R, Mehdi SH et al (2018) Antioxidative and anti-proliferative potential of
Curculigo orchioides Gaertn in oxidative stress-induced cytotoxicity: in vitro, ex vivo and
in silico studies. Food Chem Toxicol 115:244–259. https://doi.org/10.1016/j.fct.2018.03.013
100. Bhummaphan N, Petpiroon N, Prakhongcheep O et al (2019) Lusianthridin targeting of
lung cancer stem cells via Src-STAT3 suppression. Phytomedicine 62:152932. https://doi.
org/10.1016/j.phymed.2019.152932
101. Nelson VK, Sahoo NK, Sahu M et al (2020) In vitro anticancer activity of Eclipta alba whole
plant extract on colon cancer cell HCT-116. BMC Complement Med Ther 20:355. https://doi.
org/10.1186/s12906-020-03118-9
102. Sulistyani N, Nurkhasanah N (2017) The cytotoxic effect of Elephantopus scaber Linn
extract against breast cancer (T47D) cells. International Pharmacy Conference. IOP Conf
Ser: Mater Sci Eng 259:012006. https://doi.org/10.1088/1757-899X/259/1/012006
103. Liberal J, Costa G, Carmo A et al (2019) Chemical characterization and cytotoxic potential
of an ellagitannin-enriched fraction from Fragaria vesca leaves. Arab J Chem 12:3652–3666.
https://doi.org/10.1016/J.ARABJC.2015.11.014
104. Kardan M, Yazdani Z, Morsaljahan Z et al (2019) Cytotoxic effect of methanolic extracts of
Fritillaria imperialis bulbs and Eryngium caucasicum leaves on hepatoma and colon cancer
cells. Asian Pac J Trop Biomed 9(8):353–358. https://doi.org/10.4103/2221-1691.262084
1170 P. Soni et al.
105. Chen Z, Li W, Santhanam RK et al (2019) Bioactive peptide with antioxidant and anticancer
activities from black soybean [Glycine max (L.) Merr.] byproduct: isolation, identification
and molecular docking study. Eur Food Res Technol 245:677–689. https://doi.org/10.1007/
s00217-018-3190-5
106. Wang Y, Liu L, Ji F et al (2018) Soybean (Glycine max) prevents the progression of breast
cancer cells by downregulating the level of histone demethylase JMJD5. J Cancer Res Ther
14(Supplement):S609–S615. https://doi.org/10.4103/0973-1482.187292
107. Kwak Y, Ju J (2017) Glycine max Merr. leaf extract possesses anti-oxidant properties,
decreases inflammatory mediator production in murine macrophages, and inhibits growth,
migration, and adhesion in human cancer cells. Food Sci Biotechnol 26(1):245–253. https://
doi.org/10.1007/s10068-017-0033-2
108. Zheng C, Han L, Wu S (2019) A metabolic investigation of anticancer effect of G. glabra
root extract on nasopharyngeal carcinoma cell line, C666-1. Mol Biol Rep 46(4):3857–3864.
https://doi.org/10.1007/s11033-019-04828-1
109. Packialakshmi B, Sowndriya SR (2019) Anti-cancer effect of Gymnema sylvestre leaf extract
against MG63, human osteosarcoma cell line - an in vitro analysis. Int J Curr Res Rev 11(11).
https://doi.org/10.31782/IJCRR.2019.11114
110. Hejazi II, Khanam R, Mehdi SH et al (2017) New insights into the antioxidant and apoptotic
potential of Glycyrrhiza glabra L. during hydrogen peroxide-mediated oxidative stress: an in
vitro and in silico evaluation. Biomed Pharmacother 94:265–279. https://doi.org/10.1016/j.
biopha.2017.06.108
111. Li Y, Huang J, Lin W et al (2016) In vitro anticancer activity of a nonpolar fraction from
Gynostemma pentaphyllum (Thunb.) Makino. Evid Based Complement Alternat Med
2016:6308649. https://doi.org/10.1155/2016/6308649
112. Yen CH, Lai CC, Shia TH et al (2018) Gynura divaricata attenuates tumor growth and
tumor relapse after cisplatin therapy in HCC xenograft model through suppression of cancer
stem cell growth and Wnt/β-catenin signalling. J Ethnopharmacol 213:366–375. https://doi.
org/10.1016/j.jep.2017.07.019
113. Nguyen C, Baskaran K, Pupulin A et al (2019) Hibiscus flower extract selectively induces
apoptosis in breast cancer cells and positively interacts with common chemotherapeutics.
BMC Complement Altern Med 19(1):98. https://doi.org/10.1186/s12906-019-2505-9
114. Moodley S, Dwarka D, Baijnath H et al (2021) In vitro anti-cancer, anti-hypertensive and
anti-hyperglycaemic effect of Hypoxis colchicifolia. J Nat Remedies 21(3). https://doi.
org/10.18311/jnr/2021/25315
115. Keshava R, Muniyappa N, Gope R (2020) Bioactivity-guided fractionation and elucida-
tion of anti-cancer properties of Imperata cylindrica leaf extracts. Asian Pac J Cancer Prev
21(3):707–714. https://doi.org/10.31557/APJCP.2020.21.3.707
116. Asep S, Hening H, Gema SP et al (2017) Anticancer activity of Jatrophone an isolated com-
pound from Jatropha gossypifolia plant against hepatocellular cancer cell hep G2 1886.
Biomed Pharmacol J 10(2). https://doi.org/10.13005/bpj/1154
117. Potikanond S, Sookkhee S, Na Takuathung M et al (2017) Kaempferia parviflora extract
exhibits anti-cancer activity against HeLa cervical cancer cells. Front Pharmacol 8:630.
https://doi.org/10.3389/fphar.2017.00630
118. Ishteyaque S, Mishra A, Mohapatra S et al (2020) In vitro: cytotoxicity, apoptosis and ame-
liorative potential of Lawsonia inermis extract in human lung, colon and liver cancer cell line.
Cancer Investig 38(8–9):476–485. https://doi.org/10.1080/07357907.2020.1811300
119. Mohan A, Nair SV, Lakshmanan VK (2017) Leucas aspera nanomedicine shows superior
toxicity and cell migration retarded in prostate cancer cells. Appl Biochem Biotechnol
181(4):1388–1400. https://doi.org/10.1007/s12010-016-2291-5
120. Tannous S, Haykal T, Dhaini J et al (2020) The anti-cancer effect of flaxseed lignan deriva-
tives on different acute myeloid leukemia cancer cells. Biomed Pharmacother 132:110884.
https://doi.org/10.1016/j.biopha.2020.110884
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1171
121. Ma Y, Yan F, Wei W et al (2020) Litchi seed aqueous extracts play a role in suppression of
epithelial-mesenchymal transition, invasion and migration in breast cancer cells. Cell Cycle
19(3):317–325. https://doi.org/10.1080/15384101.2019.1710912
122. Lauricella M, Lo Galbo V, Cernigliaro C et al (2019) The anti-cancer effect of Mangifera
indica L. peel extract is associated to γH2AX-mediated apoptosis in colon cancer cells.
Antioxidants (Basel) 8(10):422. https://doi.org/10.3390/antiox8100422
123. Pumiputavon K, Chaowasku T, Saenjum C et al (2017) Cell cycle arrest and apoptosis induc-
tion by methanolic leaves extracts of four Annonaceae plants. BMC Complement Altern Med
17(1):294. https://doi.org/10.1186/s12906-017-1811-3
124. Dadashpour M, Firouzi-Amandi A, Pourhassan-Moghaddam M et al (2018) Biomimetic syn-
thesis of silver nanoparticles using Matricaria chamomilla extract and their potential antican-
cer activity against human lung cancer cells. Mater Sci Eng C Mater Biol Appl 92:902–912.
https://doi.org/10.1016/j.msec.2018.07.053
125. Khan F, Pandey P, Ahmad V et al (2020) Moringa oleifera methanolic leaves extract induces
apoptosis and G0/G1 cell cycle arrest via downregulation of Hedgehog Signaling Pathway
in human prostate PC-3 cancer cells. J Food Biochem 44(8):e13338. https://doi.org/10.1111/
jfbc.13338
126. Madi N, Dany M, Abdoun S et al (2016) Moringa oleifera’s nutritious aqueous leaf extract
has anticancerous effects by compromising mitochondrial viability in an ROS-dependent
manner. J Am Coll Nutr 35(7):604–613. https://doi.org/10.1080/07315724.2015.1080128
127. Erden Y (2021) Sour black mulberry (Morus nigra L.) causes cell death by decreasing mutant
p53 expression in HT-29 human colon cancer cells. Food. Bioscience 42:101113. https://doi.
org/10.1016/j.fbio.2021.101113
128. Turan I, Demir S, Kilinc K et al (2017) Antiproliferative and apoptotic effect of Morus
nigra extract on human prostate cancer cells. Saudi Pharm J 25(2):241–248. https://doi.
org/10.1016/j.jsps.2016.06.002
129. Sinha S, Sharma S, Vora J et al (2018) Mucuna pruriens (L.) DC chemo sensitizes human
breast cancer cells via downregulation of prolactin-mediated JAK2/STAT5A signaling. J
Ethnopharmacol 217:23–35. https://doi.org/10.1016/j.jep.2018.02.006
130. Aniqa A, Kaur S, Sadwal S (2021) A review of the anti-cancer potential of Murraya koenigii
(Curry Tree) and its active constituents. Nutr Cancer 74:12. https://doi.org/10.1080/0163558
1.2021.1882509
131. Utaipan T, Athipornchai A, Suksamrarn A et al (2017) Isomahanine induces endoplasmic
reticulum stress and simultaneously triggers p38 MAPK-mediated apoptosis and autophagy in
multidrug-resistant human oral squamous cell carcinoma cells. Oncol Rep 37(2):1243–1252.
https://doi.org/10.3892/or.2017.5352
132. Ju A, Cho YC, Kim BR et al (2018) Anticancer effects of methanol extract of Myrmecodia
platytyrea Becc. leaves against human hepatocellular carcinoma cells via inhibition of
ERK and STAT3 signaling pathways. Int J Oncol 52(1):201–210. https://doi.org/10.3892/
ijo.2017.4178
133. Bashari M, Yuniarti E, Putri T et al (2020) Inhibition capacity of the n-hexane fraction
of Myrmecodia pendens as a potential anti-cancer in breast and cervical cancer: in vitro
study. Indones J Cancer Chemoprevention 11(3):115–123. https://doi.org/10.14499/
indonesianjcanchemoprev11iss3pp115-123
134. Bashari MH, Hidayat S, Ruswandi YSR et al (2018) The n-hexane fraction of Myrmecodia
pendans inhibits cells survival and proliferation in colon cancer cell line. Int J Pharm Pharm
Sci 10(1):108. https://doi.org/10.22159/IJPPS.2018V10I1.21882
135. Yuletnawati SE, Meiyanto E, Agustina D (2016) High antitumor activity of ethanolic extracts
of papua’s ant nest plant (Myrmecodia tuberosa) on an oral carcinoma (KB) cell line. Int J Sci
Res 5:1619–1623. https://doi.org/10.21275/v5i1.nov1531391
136. Wu CH, Yang MY, Lee YJ et al (2017) Nelumbo nucifera leaf polyphenol extract inhibits
breast cancer cells metastasis in vitro and in vivo through PKCα targeting. J Funct Foods
37:480–490. https://doi.org/10.1016/j.jff.2017.08.021
1172 P. Soni et al.
137. Khurshid Y, Syed B, Simjee SU et al (2020) Antiproliferative and apoptotic effects of pro-
teins from black seeds (Nigella sativa) on human breast MCF-7 cancer cell line. BMC
Complement Med Ther 20:5. https://doi.org/10.1186/s12906-019-2804-1
138. Harsha M, Mohan Kumar KP, Kagathur S et al (2020) Effect of Ocimum sanctum extract
on leukemic cell lines: a preliminary in-vitro study. J Oral Maxillofac Pathol 24(1):93–98.
https://doi.org/10.4103/jomfp.JOMFP_181_19
139. Yadav R, Saini H, Kumar D et al (2019) Bioengineering of Piper longum L. extract-mediated
silver nanoparticles and their potential biomedical applications. Mater Sci Eng C Mater Biol
Appl 104:109984. https://doi.org/10.1016/j.msec.2019.109984
140. Maheswari P, Harish S, Ponnusamy S et al (2021) A novel strategy of nanosized herbal
Plectranthus amboinicus, Phyllanthus niruri and Euphorbia hirta treated TiO2 nanoparticles
for antibacterial and anticancer activities. Bioprocess Biosyst Eng 44(8):1593–1616. https://
doi.org/10.1007/s00449-020-02491-6
141. Li W, Zhang Q, Chen K et al (2019) 2-Ethoxystypandrone, a novel small-molecule STAT3
signaling inhibitor from Polygonum cuspidatum, inhibits cell growth and induces apoptosis
of HCC cells and HCC Cancer stem cells. BMC Complement Altern Med 19:38. https://doi.
org/10.1186/s12906-019-2440-9
142. Li L, Thakur K, Cao YY et al (2020) Anticancerous potential of polysaccharides sequentially
extracted from Polygonatum cyrtonema Hua in Human cervical cancer Hela cells. Int J Biol
Macromol 148:843–850. https://doi.org/10.1016/j.ijbiomac.2020.01.223
143. Singh C, Anand SK, Tiwari KN et al (2021) Phytochemical profiling and cytotoxic evaluation
of Premna serratifolia L against human liver cancer cell line. 3 Biotech 11(3):115. https://
doi.org/10.1007/s13205-021-02654-6
144. Balkrishna A, Sharma VK, Das SK et al (2020) Characterization and anti-cancerous effect of
Putranjiva roxburghii seed extract-mediated silver nanoparticles on human colon (HCT-116),
pancreatic (PANC-1) and breast (MDA-MB 231) cancer cell lines: a comparative study. Int J
Nanomedicine 15:573–585. https://doi.org/10.2147/IJN.S230244
145. Hajrah NH, Abdul WM, Al-Garni SM et al (2019) Gene expression profiling to elucidate
the pharmacological and toxicological effects of Ricinus communis L. leaf extract in mam-
malian cells. Biotechnol Biotechnol Equip 33(1):397–407. https://doi.org/10.1080/1310281
8.2019.1578691
146. Ansari S, Hasan K, Bhat S (2021) Anticancer, antioxidant, and hepatoprotective activity
of Saussurea lappa, C.B. clarke (qust) on human hepatoma cell line. J Cancer Res Ther
17(2):499–503. https://doi.org/10.4103/jcrt.JCRT_571_19
147. Ndlovu MJ, Bagla VP, Mokgotho MP et al (2021) Potential anticancer activity of Acetone
extracts of Toona cilliata, Seriphium plumosum and Schkuhria pinnata on HeLa cervical
cancer cells. Afr Health Sci 21(2):663–672. https://doi.org/10.4314/ahs.v21i2.23
148. Nawaz A, Jamal A, Arif A et al (2021) In vitro cytotoxic potential of Solanum nigrum
against human cancer cell lines. Saudi J Biol Sci 28(8):4786–4792. https://doi.org/10.1016/j.
sjbs.2021.05.004
149. Lai YJ, Tai CJ, Wang CW et al (2016) Anti-cancer activity of Solanum nigrum (AESN)
through suppression of mitochondrial function and epithelial-mesenchymal transition (EMT)
in breast cancer cells. Molecules 21(5):553. https://doi.org/10.3390/molecules21050553
150. Koh RY, Lim FP, Ling LSY et al (2017) Anticancer mechanisms of Strobilanthes crispa
Blume hexane extract on liver and breast cancer cell lines. Oncol Lett 14(4):4957–4964.
https://doi.org/10.3892/ol.2017.6821
151. Li L, Mangali S, Kour N et al (2021) Syzygium cumini (jamun) fruit-extracted phytochemicals
exert anti-proliferative effect on ovarian cancer cells. J Cancer Res Ther 17(6):1547–1551.
https://doi.org/10.4103/jcrt.JCRT_210_20
152. Khodavirdipour A, Zarean R, Safaralizadeh R (2021) Evaluation of the anti-cancer effect
of Syzygium cumini ethanolic extract on HT-29 colorectal cell line. J Gastrointest Cancer
52(2):575–581. https://doi.org/10.1007/s12029-020-00439-3
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1173
170. Abedini MR, Erfanian N, Nazem H et al (2016) Anti-proliferative and apoptotic effects of
Ziziphus Jujube on cervical and breast cancer cells. Avicenna J Phytomed 6(2):142–148
171. Kumar U, Sharma U, Rathi G (2017) Reversal of hypermethylation and reactivation of
glutathione S-transferase pi 1 gene by curcumin in breast cancer cell line. Tumour Biol
39(2):1010428317692258. https://doi.org/10.1177/1010428317692258
172. Liu Y, Zhou J, Hu Y et al (2017) Curcumin inhibits growth of human breast cancer cells
through demethylation of DLC1 promoter. Mol Cell Biochem 425(1–2):47–58. https://doi.
org/10.1007/s11010-016-2861-4
173. Chen CQ, Yu K, Yan QX et al (2013) Pure curcumin increases the expression of SOCS1 and
SOCS3 in myeloproliferative neoplasms through suppressing class I histone deacetylases.
Carcinogenesis 34(7):1442–1449. https://doi.org/10.1093/carcin/bgt070
174. Lee SJ, Krauthauser C, Maduskuie V et al (2011) Curcumin-induced HDAC inhibition and
attenuation of medulloblastoma growth in vitro and in vivo. BMC Cancer 11:144. https://doi.
org/10.1186/1471-2407-11-144
175. Dou H, Shen R, Tao J et al (2017) Curcumin suppresses the colon cancer proliferation by
inhibiting Wnt/β-catenin pathways via miR-130a. Front Pharmacol 8:877. Published 2017
Nov 24. https://doi.org/10.3389/fphar.2017.00877
176. Sohn EJ, Bak KM, Nam YK et al (2018) Upregulation of microRNA 344a-3p is involved in
curcumin-induced apoptosis in RT4 schwannoma cells. Cancer Cell Int 18:199. https://doi.
org/10.1186/s12935-018-0693-x
177. Meng J, Tong Q, Liu X et al (2017) Epigallocatechin-3-gallate inhibits growth and induces
apoptosis in esophageal cancer cells through the demethylation and reactivation of the p16
gene. Oncol Lett 14:1152–1156. https://doi.org/10.3892/ol.2017.6248
178. Ciesielski O, Biesiekierska M, Balcerczyk A (2020) Epigallocatechin-3-gallate (EGCG) alters
histone acetylation and methylation and impacts chromatin architecture profile in human
endothelial cells. Molecules 25(10):2326. https://doi.org/10.3390/molecules25102326
179. Lee YH, Kwak J, Choi HK et al (2012) EGCG suppresses prostate cancer cell growth modu-
lating acetylation of androgen receptor by anti-histone acetyltransferase activity. Int J Mol
Med 30(1):69–74. https://doi.org/10.3892/ijmm.2012.966
180. Lin CH, Wang HH, Chen TH et al (2020) Involvement of microRNA-296 in the inhibi-
tory effect of epigallocatechin gallate against the migratory properties of anoikis-resistant
nasopharyngeal carcinoma cells. Cancers (Basel) 12(4):973. https://doi.org/10.3390/
cancers12040973
181. Zhu Y, Huang Y, Liu M et al (2019) Epigallocatechin gallate inhibits cell growth and regu-
lates miRNA expression in cervical carcinoma cell lines infected with different high-risk
human papillomavirus subtypes. Exp Ther Med 17:1742–1748. https://doi.org/10.3892/
etm.2018.7131
182. Sanaei M, Kavoosi F, Roustazadeh A et al (2018) Effect of genistein in comparison with
trichostatin A on reactivation of DNMTs genes in hepatocellular carcinoma. J Clin Transl
Hepatol 6(2):141–146. https://doi.org/10.14218/JCTH.2018.00002
183. Bilir B, Sharma NV, Lee J et al (2017) Effects of genistein supplementation on genome-wide
DNA methylation and gene expression in patients with localized prostate cancer. Int J Oncol
51(1):223–234. https://doi.org/10.3892/ijo.2017.4017
184. Majid S, Dar AA, Shahryari V et al (2010) Genistein reverses hypermethylation and induces
active histone modifications in tumor suppressor gene B-Cell translocation gene 3 in prostate
cancer. Cancer 116(1):66–76. https://doi.org/10.1002/cncr.24662
185. Yu Y, Xing Y, Zhang Q et al (2021) Soy isoflavone genistein inhibits hsa_circ_0031250/
miR-873-5p/FOXM1 axis to suppress non-small-cell lung cancer progression. IUBMB Life
73(1):92–107. https://doi.org/10.1002/iub.2404
186. Lynch SM, O'Neill KM, McKenna MM et al (2016) Regulation of miR-200c and miR-141
by methylation in prostate cancer. Prostate 76(13):1146–1159. https://doi.org/10.1002/
pros.23201
Anticancer Potential of Plant-Derived Compounds: An Overview of Their Epigenetic... 1175
Nikita Kale
1 Introduction
N. Kale (*)
Faculty of Pharmacy, Oriental University, Indore, India
[3]. If a successful compound has a short half-life in the circulation, its stability can
be increased enormously by encasing it within nanosized liposomes as a drug car-
rier. In the occurrence of central nervous system cancers, many drugs have strug-
gling in crossing the blood–brain barrier to strike the tumor. Drug-loaded
nanoparticles are able to perforate this barrier, and have been appear to greatly
increase therapeutic concentrations of anticancer drugs in brain tumors. The best
means to increase the usefulness and diminish the toxicity of a cancer drug is to
straight the drug to its target and conserve its concentration at the site for an ade-
quate time for therapeutic action to take consequences [4].
2 Applications of Nanotechnology
2.1 Classification of Nanoparticles
2.1.1 One-Dimension Nanoparticles
One-dimensional system, like thin film or fabricated surfaces, has been used for
decagon in electronics, chemistry, and engineering. Fabrication of thin films (sizes
1–100 nm) or monolayer is now ordinary place with in the area of solar cells or
catalysis. These thin films are using in several technological applications, including
information storage systems, chemical and biological sensors, fiber-optic systems,
magneto-optic, and optical device [5].
Disinfection
Drug screening
Advance
technologies NANOTECHNLOGY
medical imaging
Controlled
drug delivery
Drug targeting
Tissue engineering
Nanocoating
Identification, security and logistics
2.1.2 Two-Dimension Nanoparticles
2.1.3 Three-Dimension Nanoparticles
2.2 Polymeric Nanoparticles
chemical class of PNPs (e.g., van der Waals forces, hydrophobic interlinkage or
hydrogen bonding) [9].
Drugs can be crippled on PNPs surface after a polymerization reaction or can be
enclosed on PNP structure during a polymerization step. Furthermore, drugs may be
rescued by desorption, dispersal, or nanoparticle erosion in target tissue [10].
The application of biodegradable nanosystems for the enlargement of nanomedi-
cines is one of the most victorious ideas. Nanocarriers contained of biodegradable
polymers experience hydrolysis in the body, construct biodegradable metabolite
monomers, such as lactic acid and glycolic acid [11]. Negligible systemic toxicity
is correlated with using of PLGA for drug transportation or biomaterial applica-
tions. Such nanoparticles are biocompatible with tissue and cells. Drug-
biodegradable polymeric nanocarrier combined used for drug delivery is secure in
blood, nontoxic, and nonthrombogenic [12].
3 Preparation of Nanoparticles
This is one of the most commonly used methods for the preparation of nanoparti-
cles. Solvent evaporation was the first technique developed to formulate PNPs from
a solvent. In this technique, polymer solutions are prepared in volatile solvents and
emulsions are formulated. In the past, dichloromethane and chloroform preformed
polymer were extensively used, but are now restored with ethyl acetate which has a
stronger toxicological profile [14]. The emulsion is transformed into a nanoparticle
suspension on evaporation of the solvent for the polymer, which is granted to diffuse
along the continuous phase of the emulsion. In the conventional methods, two main
strategies are being used for the formation of emulsions, the preparation of single-
emulsions, e.g., oil-in-water (o/w) or double-emulsions, e.g., (water-in-oil)-in-
water, (w/o)/w. These methods utilize high-speed homogenization or ultrasonication,
followed by evaporation of the solvent, either by continuous magnetic stirring at
room temperature traditional or under decrease pressure [15]. Subsequently, the
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1181
Salting-out ground on the detachment of a water mixable solvent from watery solu-
tion via a salting-out effect. Salting-out is based on the separation of a water mis-
cible solvent from aqueous solution via a salting-out effect. Polymer and drug are
initially diffused in a solvent which is eventually emulsified into an aqueous gel
carrying the salting-out agent (electrolytes, like magnesium chloride and calcium
chloride, or nonelectrolytes like sucrose) and a colloidal stabilizer like poly vinyl
pyrrolidone or hydroxyl ethyl cellulose [17]. This oil/water emulsion is diluted with
a sufficient volume of water or aqueous solution to increase the diffusion of solvent
into the aqueous phase, thus influencing the formation of nanospheres. Various
manufacturing parameters can be diversified including stirring rate, internal/exter-
nal phase ratio, concentration of polymers in the organic phase, variety of electro-
lyte concentration, and type of stabilizer in the aqueous phase. This technique used
in the preparation of PLA, poly (methacrylic) acids, and ethyl cellulose nanospheres
shows high productivity and is easily scaled up. Salting-out does not need an
increase of temperature and consequently may be functional when heat-sensitive
substances have to be prepared. The considerable disadvantages are absolute
application to lipophilic drug and the considerable nanoparticles washing steps [18]
(Fig. 3).
3.3 Emulsion-Diffusion Method
4 Characterization of Nanoparticles
4.1 Particle Size
Particle size distribution and morphology are the principal parameters of assuming
of nanoparticles. Morphology and size are dignified by electron microscopy. The
vital application of nanoparticles is in drug release and drug targeting. It has been
found that particle size affects the drug release. Minute particles offer enormous
surface area. As a result, most of the drug stacked onto them will be disclose to the
particle surface directing to fast drug release. On the conflicting, drugs slowly dif-
fuse core huge particles. As a drawback, minute particles tend to aggregate during
storage and transportation of nanoparticle dispersion. Hence, there is a compromise
between a small size and maximum stability of nanoparticles. Polymer degradation
can also be affected by the particle size. For instance, the degradation rate of poly
(lactic-co-glycolic acid) was found to increase with increasing particle size in vitro
[25–27].
There are several tools for determining nanoparticle size as discussed below:
(a) Dynamic Light Scattering (DLS)
Currently, the quick and most popular method of regulating particle size is
photon-correlation spectroscopy (PCS) or dynamic light scattering (DLS). DLS
is widely used to dictate the size of Brownian nanoparticles in colloidal suspen-
sions in the nano and submicron ranges. Shining monochromatic light (laser)
toward a solution of spherical particles in Brownian motion causes a Doppler
shift when the light strikes the moving particle, changing the wavelength of the
arriving light. This change is connected to the size of the particle [28]. It is fea-
sible to extract the size distribution and offer a description of the particle is
shifting in the medium, estimating the diffusion coefficient of the particle and
using the auto connection function. The photon correlation spectroscopy (PCS)
represents the most regularly used technique for correct estimation of the par-
ticle size and size distribution based on DLS [29].
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1185
4.2 Surface Charge
The nature and toughness of the surface charge of nanoparticles are extremely
important as it investigates their interlinkage with the biological environment as
well as their electrostatic correlation with bioactive hybrid. The colloidal stability is
examined through zeta potential of nanoparticles. This potential is a divergent esti-
mate of the surface charge. It corresponds to potential variation between the exter-
nal Helmholtz plane and the surface of shear. The measurement of the zeta potential
grants for predictions about the storage stability of colloidal dispersion. High zeta
potential values, likewise positive or negative, must be accomplished in order to
certify stability and avoid accumulation of the particles. The level of surface hydro-
phobicity can then be forecasted from the ethics of zeta potential. The zeta potential
can also contribute information concerning the nature of material enclosed within
the nanocapsules or coated onto the surface [35].
4.3 Surface Hydrophobicity
1. Enlarging the effectiveness of drug delivery and diminishing side effects, con-
sequently toxicity
2. Particular targeting of the dynamic components in cell/tissues
3. Boosting the properties of pharmacologically vital drugs like stability, solubil-
ity, half-life, and tumor accumulation
4. Producing stimuli-responsive drug deliver
5. Enlarging the area of drugs enclosed/connected to biomacromolecules like pro-
teins, mRNA
6. Development of therapeutic efficiency by delivering numerous active agents to
a fixed targeted site in order to conquer hindrance such as drug resistance
7. Overcoming biological hurdle
8. Enhancing the sensitivity of diagnosis and describing of tumorous sites
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1187
The polymeric drug conveyor delivers the drug at the tissue site by any one of the
three common physicochemical mechanisms.
• By the swelling of the polymer nanoparticles by hydration accompanied by
release across diffusion
• By an enzymatic reaction arising in rupture or cleavage or debasement of the
polymer at site of delivery, thereby liberating the drug from the captured
inner core
• Segregation of the drug from the polymer and its adsorption/rescue from the
swelled nanoparticles [40]
Advantages of nanostructure-moderated drug delivery comprise the capacity to
deliver drug molecules directly into cells and the capacity to target tumors within
healthy tissue. Nanoscale drug delivery architectures are able to puncture tumors
due to the discontinuous, or “porous,” nature of the tumor microvasculature, which
usually contains pores ranging from 100 to 1000 nm in diameter. The microvascu-
lature of healthy tissue ranges by tissue type, but in most tissues comprising the
heart, brain, and lung, there are compact intercellular junctions less than 10 nm.
Through accurate control of the drug conveyor architecture, the liberate of the drug
can be adjusted to accomplish a wished kinetic profile. Three of the most ordinary
kinetic profiles are zero order, first order, and Higuchi [41, 42].
The delivery of most drugs is accomplished through oral administration or by
injection and follows first-order kinetics. The absolute release profile for most drugs
would observe a steady liberation rate so that the drug levels in the body remain
continuous while the drug is being administered. Further current transdermal drug
delivery process follows the Higuchi model. As shown eventually, nanostructured
polymeric and silica nanoparticles are being enlarged as drug carriers, which
accomplished near zero-order kinetics [43].
Zero order : Dt D0 K 0t
=
Higuchi order : Dt D=
0 KHt1 / 2
where Dt is the amount of drug released at time t, D0 is the initial amount of drug
released, result of initial quick release,
k0 is the zero-order release constant,
k1 is the first-order release constant, and
kH is the Higuchi release constant.
Liposomes (phospholipid spheres of ~100 nm in diameter that are bi-layered in
shape) are outstanding carriers for a diversity of drugs and are being examined enor-
mously for use in gene remedy protocols and targeted drug delivery in cancer. With
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1189
Fig. 6 Drug release profiles from zero-order, first-order, and Higuchi kinetics
present liposome technologies, it is not tough to transfect a cell for grounds of gene
therapy, but the therapeutic gene may be deteriorating if it is not able to traffic out
of the endosome [44]. To upgrade the efficacy of gene therapy, synthetic pH-
sensitive histidylated oligolysine can be attached to a drug-liposome complex to
support in escaping from the endosome. This protocol was appeared to enhance the
transfection efficiency in recumbent and pancreatic cancer cell lines by 39 times,
and upraised the manner of the transgene in a human prostate cancer xenograft
model in athymic nude mice without increasing toxicity [45]. Nanoparticles attached
to chemotherapeutic drugs allow them to traverse the blood–brain barrier for brain
tumor treatment. In January 2005, a nanoparticle-based drug called Abraxane
(paclitaxel protein-bound particles, Abraxis Oncology) was approved by the Food
and Drug Administration for breast cancer treatment. Abraxane uses nanoscale par-
ticles of the natural protein albumin that can be delivered in the body without the use
of solvents [46, 47] (Fig. 6).
Medicinal and structural chemists have been creating and manipulating nanome-
ter and sub nanometer-sized components of drugs for decades, and will continue to
do so into the fore salable future. The difference is that they will now be joined by
a wide variety of scientists from a number of disciplines normally not involved in
drug research [49].
A recent article written by physicists in Physical Review Letters [50], titled
“Electronic Structure and Bonding of Au on a Si Oh, not zero 2 Cluster: A Nano
bullet for Tumours,” is a good example of how research from diverse sources can
and should be brought to bear on the problem of cancer. Success in this endeavor,
however, will require a concerted effort to integrate and coordinate the research in
an approach that might now be described as “systems biology” [51, 52].
There must be an adequate drug concentration in the body to allow for an effective
dose at the tumor site. The target must be strongly inhibited, with the target’s func-
tion essential for tumor cell viability. The drug must have a high differential toxicity
toward the tumor or a favorable therapeutic window [53].
Research in nanomedicine will be addressing all these points, and a few exam-
ples in the drug development arena will be given below.
Monoclonal antibodies will be an essential component of the new wave of cancer
treatments developed through nanotechnologies. They are being used as imaging
vehicles, for drug targeting, as drug carriers, and as the drug itself. There are nine or
more FDA-approved antibodies approved for clinical use in cancer, and many more
are being evaluated in clinical trials [54]. The mechanism of action of the antibodies
includes receptor ligand-binding competition; interference with receptor function;
antibody-dependent, cell-mediated cytotoxicity; complement-dependent cellular
cytotoxicity; or, perhaps, a combination of the above. This activity can also be com-
bined with toxins directed at the cancer cell to produce an even more efficacious
drug [55]. One of the holy grails of drug research is to be able to rationally design
and produce effective small-molecule inhibitors of protein function. Development
of many drugs will be the result of application of nanotechnologies that have been
in place for many years. For example, nuclear magnetic resonance and x-ray crystal
structures of target proteins and their ligands or substrates are being used as the
template for rational design of new drugs. The target may be enzymes or receptor–
ligand proteins [56]. Inhibitors of enzymatic activity are, in general, easier to design
than blockers of protein–protein interactions. As an example, the first successful
drug approved for the treatment of chronic myeloid leukemia (CML), Gleevec-, is
an inhibitor of the tyrosine kinase mutant, BCR-ABL [57] (Table 1).
In many patients, administration of the drug results in what appears to be com-
plete remission, but Gleevec-resistant leukemia often returns through mutations in
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1191
Table 1 (continued)
S.No. Name of Plant Family Active Constituents Predicted Mode of Action
6 Luffa Cucurbitaceae Pentacyclic triterpene, Induces apoptosis by using
cylindrica bryonolic acid the mitochondrial pathway
(Linn.)M. J. rather than through the cell
Roem. cycle in ovarian cancer cell
line MDAH-2774. Cell
invasion, migration, and
colony formation capacity in
ovarian cancer cells were
reduced. Cell death was
induced by increasing
oxidative stress process in
ovarian cancer cells.
7 Symplocos Symplocaceae. Betulinic, oleanolic, The ellagic acid in the
racemosa acetyl oleanolic, and MDA-MB-231 cells showed
Roxb. ellagic acids significant antiproliferative
effects with dose-dependent
pattern. The antiproliferative
effects in MCF-7 cells were
observed in only at a high
concentration. Ellagic acid
had no effect on the cell
cycle in both breast cancer
cells.
8 Camellia Theaceae Theophylline and At micromolar
sinensis theobromine, concentrations, kaempferol
(Linn.) quercetin, kaempferol effectively inhibits the
O. Kuntze. growth of breast cancer cell
lines (VM7Luc4E2, MDA-
MB-231, MCF-7)
9 Coix Gramineae; Palmitoleic and Trans-vaccenic acid inhibits
lacryma-jobi Poaceae vaccenic acids proliferation and induces
Linn. apoptosis of human
nasopharyngeal carcinoma
cells via a mitochondrial-
mediated apoptosis pathway.
10 Crocus Iridaceae Crocin The administration of crocin
sativus Linn. leads to inhibition of cell
proliferation and also
induction of apoptosis in the
cancer cells. Crocin also
inhibits Nf-kB in
hepatocytes, suppresses S
and G2/M phases of the cell
cycle, induces apoptosis, and
down-regulates inflammation
in HepG2 cells.
(continued)
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1193
Table 1 (continued)
S.No. Name of Plant Family Active Constituents Predicted Mode of Action
11 Heliotropium Boraginaceae Indicine-Noxide Phase II studies in all panel
indicum Linn. tumors are indicated,
especially colon carcinoma
and leukemias.
12 Linum Linaceae Secoisolariciresinol Has been shown to inhibit
usitatissimum diglycoside the growth of established
Linn. human breast tumors.
13 Pastinaca Umbelliferae; Coumarins Coumarin derivatives are
sativa Linn. Apiaceae ubiquitous in nature and can
readily interact with diverse
enzymes and receptors in
cancer cells via weak bond
interactions; hence, coumarin
is a highly privileged
pharmacophore for the
development of novel
anticancer agents.
14 Plumbago Plumbaginaceae Naphthoquinone The cytotoxicity of
zeylanica derivatives, plumbagin 1,4-naphthoquinones is
Linn. related to their electron-
accepting capability, which
gives rise to ROS production
leading to DNA damage and
cancer cell apoptosis.
15 Solanum Solanaceae Beta-solamarine Induced apoptosis of these
trilobatum cells by PS externalization in
Linn. a dose-dependent manner
and increased sub-G1
fraction were observed.
Quenching of the expression
of tumor necrosis factor
receptors (TNFRs) during
the progress of human lung
carcinogenesis has been
previously reported.
16 Taxus baccata Taxaceae Diterpene amide Diterpenoid amides may be
Linn. useful in the apoptosis
induction of human leukemia
cancer cells.
17 Vinca rosea Apocynaceae Vincristine Vincristine is a
Linn. chemotherapy drug that
belongs to a group of drugs
called vinca alkaloids.
Vincristine works by
stopping the cancer cells
from separating into two new
cells. So, it stops the growth
of the cancer.
(continued)
1194 N. Kale
Table 1 (continued)
S.No. Name of Plant Family Active Constituents Predicted Mode of Action
18 Catharanthus Apocynaceae Vinblastine and The antitumor activity of
roseus (L.) Vincristine vinblastine is thought to be
G. Don. due primarily to inhibition of
mitosis at metaphase through
its interaction with tubulin.
Vinblastine binds to the
microtubular proteins of the
mitotic spindle, leading to
crystallization of the
microtubule and mitotic
arrest or cell death.
19 Lycopersicon Solanaceae N-nitrosomorpholin Decreased serum lycopene
esculentum concentrations are associated
Mill. with an increased risk of
prostate cancer.
20 Symphytum Boraginaceae Allantoin Allantoin can ameliorate
officinale cisplatin-induced neurotoxic
Linn. insults to neurons. It
therefore shows promise as
an adjuvant drug in cancer
treatment.
the active site. By careful study of the mutations and the structure of the kinase, new
small molecular inhibitors were designed that could block the mutant strains and
appear to be more efficacious than the original drug [58]. The two drugs are reported
to work synergistically, which will, hopefully, result in a complete cure of chronic
myeloid leukemia. Drugs derived from nucleic acids are beginning to make an
impact on the nanomedicine scene. Antisense technology exploits the use of oligo-
nucleotides in the range of 15 to 20 nucleotides to block the function of an RNA
target [59]. This technology has made rapid progress after experiencing initial dif-
ficulties in showing efficacy for in vivo models of disease. Ongoing clinical trials
using antisense drugs include prostate, breast, pancreatic, lung, colorectal, mela-
noma, and brain cancers [60].
A novel approach for this technology is to use oligonucleotides for sensitizing
tumor cells to chemotherapy. The oligonucleotides are being combined with nano-
liposomes to target and deliver the nucleic acids to the cancer cells and block pro-
duction of the alpha folate receptor [61]. This block was shown to decrease cell
survival of breast cancer cell lines, and sensitized a cell line by fivefold to doxoru-
bicin. This is a good example of how nanotechnologies can be used to increase the
effectiveness of existing drugs, facilitating the use of lower dosages to decrease
toxicity [62, 63].
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1195
References
1. Hahens WI, Oomen AG, de Jong WH, Cassee FR (2007) What do we (need to) know about
the kinetic properties of nanoparticles in the body? Regulatory Toxicol Pharmacol 49:217–229
2. Couvreur P, Dubernet C, Puisieux F (1995) Controlled drug delivery with nano particles:current
possibilities and future trends. Eur J Pharm Biopharm 41:2–13
3. Suri SS, Fenniri H, Singh B (2007) Nanotechnology-based drug delivery systems. J Occup
Med Toxicol 2:16
4. Hett A (2004) Nanotechnology: small matters, many unknown
5. Lachman L, Lieberman HA, Kanig JL (1999) Theory and practice of industrial pharmacy, 3rd
edn. Pharmaceutical Industry, pp 26–30
6. Tomalia DA (2004) Birth of a new macromolecular architecture: Dendrimers as quantized
building blocks for nanoscale synthetic organic chemistry. Aldrichimica Acta. 37(2):39–57
7. Wiener EC, Brechbiel MW, Brothers H, Magin RL, Gansow OA, Tomalia DA (1994)
Dendrimer-based metal chelates: a new class of magnetic resonance imaging contrast agents.
Magn Reson Med 31:1–8
8. Li Y, Cheng Y, Xu T (2007) Design, synthesis and potent pharmaceutical applications of gly-
codendrimers: a mini review. Curr Drug Discov Technol 4:246–254
9. Fu HL, Cheng SX, Zhang XZ, Zhuo RX (2007) Dendrimers/DNA complexes encapsulated in
a water-soluble polymer and supported on fast degrading star poly (DL-Lactide) for localized
gene delivery. J Control Release 124:181–188
10. Cheng Y, Wang J, Rao T, he X., Xu T. (2008) Pharmaceutical Applications of Dendrimers:
promising nanocarriers for drug delivery. Front Biosci 13:1447–1471
11. Mecke A, Uppuluri S, Sassanella TM, Lee DK, Ramamoorthy A, Baker JR Jr (2004) Direct
observation of lipid bilayer disruption by poly (amidoamine) dendrimers. Chem Phys Lipids
132:3–14
12. Larson DR, Zipfel WR, Williams RM, Clark SW, Bruchez MP, Wise FW (2003) Water-soluble
quantum dots for multiphoton fluorescence imaging in vivo. Science 300:1434–1436
13. Ghosh PK (2000) Hydrophilic polymeric nanoparticles as drug carriers. Indian J Biochem
Biophys 37:273–282
14. Prasad Rao J, Geckeler KE Polymer nanoparticles: preparation techniques and size control
parameters, progress in polymer science G model. J Pharm Pharmaceut Sci 674
15. Reis CP, Neufeld RJ, Ribeiro AJ, Veiga F, Nanoencapsulation I (2006) Methods for prepara-
tion of drug-loaded polymeric nanoparticles. Nanomed Nanotechnol Biol Med 2:8–21
16. Lemoine D, Preat V (1998) Polymeric nanoparticles as delivery system for influenza virus
glycoproteins. J Control Release 54:15–27
17. Song CX, Labhasetwar V, Murphy H, Qu X, Humphrey WR, Shebuski RJ et al (1997)
Formulation and characterization of biodegradable nanoparticles for intravascular local drug
delivery. J Control Release 43:197–212
18. Catarina PR, Ronald JN, Antonio JR (2006) Nano capsulation 1. Method of preparation of
drug – loaded polymeric nanoparticles: nano technology. Biol Med 2:8–21
19. Allemann E, Gurny R, Doekler E (1993) Drug-loaded nanoparticles preparation methods and
drug targeting issues. Eur J Pharm Biopharm 39:173–191
20. Quintanar-Guerrero D, Allemann E, Fessi H, Doelker E (1998) Preparation techniques and
mechanism of formation of biodegradable nanoparticles from preformed polymers. Drug Dev
Ind Pharm 24:1113–1128
21. Jung T, Kamm W, Breitenbach A, Kaiserling E, Xiao JK, Kissel T (2000) Biodegradable nano
particles for oral delivery of peptides: is there a role for polymer to affect mucosal uptake? Eur
J Pharm Biopharm 50:147–160
22. Vargas A, Pegaz B, Devefve E, Konan-Kouakou Y, Lange N, Ballini JP (2004) Improved pho-
todynamic activity of porphyrin loaded into nano particles: an in vivo evaluation using chick
embryos. Int J Pharm 286:131–145
1196 N. Kale
23. Yoo HS, Oh JE, Lee KH, Park TG (1999) Biodegradable nanoparticles containing PLGA con-
jugates for sustained release. Pharm Res 16:1114–1118
24. El-Shabouri MH (2002) Positively charged nano particles for improving the oral bioavailabil-
ity of cyclosporine-a. Int J Pharm 249:101–108
25. Chorney M, DAneuberg H, Golomb G (2002) Lipophilic drug-loaded nanospheres by nano
precipitation: effect of the formulation variables on size, drug recovery and release kinetics. J
Control Releas E 83:389–400
26. Betancor L, Luckarift HR (2008) Trends Biotechnol 26:566
27. Venkatesan P, SreeJanardha Nan V, Muralidharan C, Valliappan K (2012) Improved HPLC
method with the aid of chemometric strategy: determination of loxoprofen in pharmaceutical
formulation. Acta Chim Slov 59:242–248
28. Venkatesan P, Manavalan R, Valliappan K Preparation and evaluation of sustained release
loxoprofen-loaded microspheres. J Basic Clin Pharm
29. De Assis DN, Mosqueira VC, Vilela JM, Andrade MS, Cardoso VN (2008) Release profiles
and morphological characterization by atomic force microscopy and photon correlation spec-
troscopy of 99 m technetium – fluconazole nanocapsules. Int J Pharm 349:152–160
30. Molpeceres J, Aberturas MR, Guzman M (2000) Biodegradable nanoparticles as a delivery
system for cyclosporine: preparation and characterization. J Microencapsul 17:599–614
31. Venkatesan P et al (2011) Int J Pharm Biomed Res (IJPBR) 2(3):107–117
32. Sathiya Sundar R, Murugesan A, Venkatesan P, Manavalan R (2010) Formulation development
and evaluation of carprofen microspheres. Int J Pharm Tech Res 2(3):1674–1676
33. Soppinath KS, Aminabhavi TM, Kulkurni AR, Rudzinski WE (2001) Biodegradable poly-
meric nanoparticles as drug delivery devices. J Control Release 70:1–20
34. Polakovic M, Gorner T, Gref R, Dellacherie E (1999) Lidocaine-loaded biodegradable nano-
spheres. II. Modelling of drug release. J Control Release. 60:169–177
35. Pangi Z, Beletsi A, Evangelatos K (2003) PEG-ylated nanoparticles for biological and phar-
maceutical application. Adv Drug Del Rev 24:403–419
36. Scholes PD, Coombes AG, Illum L, Davis SS, Wats JF, Ustariz C, Vert M, Davies MC (1999)
Detection and determination of surface levels of poloxamer and pva surfactant on biodegrad-
able nanospheres using SSIMS and XPS. J Control Release 59:261–278
37. Shi J, Kantoff PW, Wooster R, Farokhzad OC (2016) Cancer nanomedicine: Progress, chal-
lenges and opportunities. Nat Rev Cancer 17:20. [CrossRef] [PubMed]
38. Sahoo SK et al (2003) Nanotech approaches to drug delivery and imaging. Drug Discov Today
8:1112–1120
39. Vasir JK et al (2005) Nanosystems in drug targeting: opportunities and challenges. Curr
Nanosci 1:47–64
40. Kipp JE (2004) The role of solid nanoparticle technology in the parenteral delivery of poorly
water-soluble drugs. Int J Pharm 284:109–122
41. Rabinow BE (2004) Nanosuspensions in drug delivery. Nat Rev Drug Discov 3:785–796
42. Horn D, Rieger J (2001) Organic nanoparticles in the aqueous phase-theory, experiment, and
use. Angew Chem Int Ed 40:4330–4361
43. Torchilin VP (2005) Recent advances with liposomes as pharmaceutical carriers. Nat Rev
Drug Discov 4:145–160
44. Wissing SA, Kayser O, Muller RH (2004) Solid lipid nanoparticles for parenteral drug deliv-
ery. Adv Drug Deliv Rev 56:1257–1272
45. Koziara JM et al (2004) Paclitaxel nanoparticles for the potential treatment of brain tumors. J
Control Release 99:259–269
46. Steiniger SC et al (2004) Chemotherapy of glioblastoma in rats using doxorubicin-loaded
nanoparticles. Int J Cancer 109:759–767
47. Brannon-Peppas L, Blanchette JO (2004) Nanoparticle and targeted systems for cancer ther-
apy. Adv Drug Delivery Rev 56:1649–1659
48. Hood JD et al (2002) Tumor regression by targeted gene delivery to the neovasculature.
Science 296:2404–2407
Treatment of Cancer Using Combination of Herbal and Novel Drug Delivery System 1197
49. Whitesides GM (2003) The “Right” size in nanobiotechnology. Nat Biotechnol 21:1161–1165
50. Sun Q, Wang Q, Rao BK, Jena P (2004) Electronic structure and bonding of au on a SiO2
cluster: a nanobullet for tumors. Phys Rev Lett 93:186803/1–186,803/4
51. Hood L, Heath JR, Phelps ME, Lin B (2004) Systems biology and new technologies enable
predictive and preventative medicine. Science 306(2004):640–643
52. Hood L, Perlmutter RM (2004) The impact of systems approaches on biological problems in
drug discovery. Nat Biotechnol 22:1215–1217
53. Kamb A (2005) Opinion: what’s wrong with our cancer models? Nat Rev Drug Discov
4:161–165
54. Ferrara N, Hillan KJ, Gerber HP, Novotny W (2004) Discovery and development of bevaci-
zumab, an Anti- VEGF antibody for treating cancer. Nat Rev Drug Discov 3:391–400
55. Groner HC, Wels W (2004) Therapeutic antibodies. Curr Mol Med 4:539–547
56. Von Eschenbach AC (2004) A vision for the national cancer program in the United States. Nat.
Rev. Cancer 4:820–828
57. Arkin MR, Wells JA (2004) Small-molecule inhibitors of protein-protein interactions: pro-
gressing towards the dream. Nat Rev Drug Discov 3:301–317
58. Brooijmans N, Kuntz ID (2003) Molecular recognition and docking algorithms. Annu Rev
Biophys Biomol Struct 32:335–373
59. Card GL, Blasdel L, England BP, Zhang C, Suzuki Y, Gillette Y (2005) A family of phos-
phodiesterase inhibitors discovered by cocrystallography and scaffold-based drug design. Nat
Biotechnol 23:201–207
60. Erlanson DA, Wells JA, Braisted AC (2004) Tethering: fragment-based drug discovery. Annu
Rev Biophys Biomol Struct 33:199–223
61. Lipinski C, Hopkins A (2004) Navigating chemical space for biology and medicine. Nature
432:855–861
62. Rees DC, Congreve M, Murray CW, Carr R (2004) Fragment-based lead discovery. Nat Rev
Drug Discov 3:660–672
63. Shoichet BK (2004) Virtual screening of chemical libraries. Nature 432:862–865
64. Khare CP (2007) Indian Medicinal Plants, Khare CP ed, Published by Springer,
pp. 103,115,163,165,171 &179
Anti-inflammatory Potential of Lead
Compounds and Their Derivatives
from Medicinal Plants
1 Introduction
Since the late eighteenth century, the inflammatory process has been reported as one
of the most attractive areas of epidemiology and pharmacy. The word inflammation
is derived from the Latin word “Inflammatio” which is a complex cascade of reac-
tions organized by immune cells, molecular mediators, etc. that tend to respond to
injurious agents which can be chronic or acute. The five classic hallmark signs of
inflammation include heat, pain, redness, swelling, and loss of function [1].
Inflammation starts with an acute reaction and enters into prolonged or chronic
phase when left untreated (Fig. 1). Chronic inflammation can lead to various conse-
quences including arthritis [2], asthma [3], atherosclerosis [4], autoimmune dis-
eases [5], diabetes [6], and cancer [7].
Over a decade, inflammatory diseases seem to represent a chief cause of morbid-
ity [8]. Current therapy for inflammatory diseases is majorly confined to the usage
of steroidal and nonsteroidal anti-inflammatory agents. The chronic use of these
drugs is reported to in turn cause severe adverse effects on gastrointestinal, cardio-
vascular, renal functions [9, 10]. These adverse effects of available anti-inflammatory
drugs lead to the need for mandatory knowledge on molecular mechanism behind
inflammation and explore drugs or bioactive compounds with mere or no toxicity.
Natural sources such as plants and their phytochemicals, microbes/animals, and
their bioactive elements are found to be promising sources of new anti-inflammatory
agents. Herbal medicine, Phytomedicine, and Siddha medicine are recently gaining
momentum throughout the world due to the fact that they are safe, effective, and
cheaper than conventional modern medicines. Herbs and plants have been used to
treat human diseases since ancient times. According to Rig Veda which dates back
to 4500–1600 BC, medicinal plants were used to treat many human ailments [11].
In modern medicine, more than 200 compounds of plant origin have been commer-
cialized. Plant parts such as roots (Withania somnifera), leaves (Aegle marmelos),
fruits (Morinda citrifolia), etc. are employed as source of medicines [12, 13]. Some
of the important bioactive compounds of plants include alkaloids, flavonoids, tan-
nins, and phenolic compounds [14]. These bioactive compounds also hold antidia-
betic, antibacterial, antithrombotic, hypotensive, anti-inflammatory, and anti-cancer
potential. Although various in vitro and in vivo models for anti-inflammatory drug
development are available, systematic evaluation of phytoconstituents can facilitate
the identification and development of potential anti-inflammatory lead molecules
from natural sources. This chapter focuses on typical inflammatory diseases, its
molecular targets, NSAIDs in current usage, advantages of plant lead compounds
over NSAIDs with primary focus on the herbal plants, phytochemical classes, and
lead molecules with potent anti-inflammatory potential.
2 Inflammatory Diseases
Dermal inflammatory diseases are one among the inflammatory diseases that occur
in the skin and other dermal tissues. This disease may occur at three levels viz.,
epidermis, dermis, and subcutaneous layer. According to Brinster Classification
[25], dermal or skin inflammatory diseases are of eight types viz., bullous, interface,
nodular and diffuse, spongiotic, vasculitis, perivascular, panniculitis, and psoriasi-
form. In bullous pemphigoid inflammation, the immune system produces antibodies
to the fibers that connect the epidermis and the dermis. These antibodies trigger
inflammation that produces the blisters and itching of bullous pemphigoid [26]. In
case of psoriasiform, the condition resembles the psoriasis [27].
Based on the location of oral mucosa, dental inflammations are classified as glos-
sitis, stomatitis, gingivitis, periodontitis or cheilitis. If deeper structures are involved,
it might be a phlegmon or abscess [38]. Glossitis is mean soreness of the tongue, or
more usually inflammation with depapillation of the dorsal surface of the tongue
(loss of the lingual papillae) [39, 40]. It can be acute or chronic. The term Stomatitis
is derived from the Greek word stoma meaning “mouth” and the suffix -itis meaning
“inflammation.” It refers to any inflammatory process affecting the mucous mem-
branes of the mouth and lips, with or without oral ulceration [41].
In early stages of gingivitis, the gums become swollen, red, and may bleed. It is
considered the main cause of tooth loss for adults worldwide [42, 43]. Later or seri-
ous form of gingivitis is called as periodontitis, where the gums can pull away from
the tooth, bone can be lost, and the teeth may loosen or fall out. Cheilitis are inflam-
mation in lips and may be acute or chronic inflammation with chapped lips [44].
predominantly by bacteria [52]. Symptoms include blood and pus in the urine, pain
during urination, pain in back and sides, and urgent or infrequent urination [53, 54].
Renal inflammation can affect other organs besides the kidneys as part of an autoim-
mune disorder. Lupus erythematosus is one of the well-known renal inflammatory
diseases [55].
Respiratory inflammatory diseases occur in the respiratory tract of the human body
could be either acute or chronic. Infection with pneumonia and acute respiratory
distress syndrome (ARDS) are taken into account as acute, whereas chronic inflam-
mation is represented by asthma and chronic obstructive pulmonary disease (COPD)
[56]. Some well-known examples for respiratory inflammation include sinusitis,
pharyngitis, epiglottitis, laryngotracheitis, and bronchitis. Sinusitis is known as the
inflammation of mucosal membrane that lines the sinuses [57]. Bronchitis is identi-
fied by the inflammation of bronchi, the airways of lungs. Symptoms of respiratory
inflammatory diseases include coughing, production of sputum, chest pain, and
shortness of breath. Epiglottitis is a condition wherein inflammation and edema of
epiglottis and neighboring structures such as arytenoids, aryepiglottic folds, and
vallecula occur that blocks the entry of air into lungs [58]. It is frequently caused by
the bacteria Haemophilus influenzae type B (HIB) in children [59].
3.1 Mechanism of Action
NSAIDs act by inhibiting the cyclooxygenase enzymes COX-1 and COX-2, thereby
blocking the synthesis of prostaglandins. COX-1 is constitutively expressed in
healthy cells and it is responsible for maintaining mucosal barrier in the GI tract,
renal hemostasis, and platelet aggregation whereas, COX-2 is expressed only during
pain and inflammation [60].
1204 N. S. Nirmala et al.
Tissue damage leads to the release of various cytokines, hormones, and other
substances which in turn activate phospholipase A2 (Fig. 3). As a result, arachidonic
acid is released from membrane lipids. The two cyclooxygenase isoforms COX-1
and COX-2 are required to convert the arachidonic acid into unstable, prostaglandin
G2 (PGG2) which is further converted to a more stable form, prostaglandin H2
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1205
NSAIDs are keystone in therapy against chronic and acute pain. NSAIDs have anti-
inflammatory, analgesic, and antipyretic activity, available in the parenteral, topical,
intramuscular, and rectal forms. Generally, after a minor or major surgery or tooth
extraction, most of the patients will suffer from postoperative pain through the gen-
eration of prostaglandins. NSAIDs are used to reduce pain and inflammation alone
or in combination with opioids, local analgesics, and or adjuvants [61]. Some of the
ailments /disease with recommended NSAIDs are tabulated (Table 1).
3.3 Disadvantages of NSAIDs
NSAIDs show a ceiling effect where the increase of dose increases only the side
effects but not the therapeutic effect [61]. Continuous use of NSAIDs leads to the
inhibition of prostaglandins and destroys the GI mucosal barrier, which leads to GI
bleeding [68]. People with asthma are easily prone to adverse effects of NSAIDs.
Diclofenac causes the majority of the NSAIDs-related cardiovascular side effects
[60]. COX-2 inhibitor shows very less side effects when compared to nonselective
COX inhibitor. Some of the major side effects of NSAIDs and its pathophysiologi-
cal conditions are tabulated (Table 2).
Table 2 List of major side effects of NSAIDs and its pathophysiological conditions
Side effects Pathophysiology condition References
Renal side effects Fluid-electrolyte disorders [60]
Renal papillary necrosis [68]
Acute renal dysfunction
Interstitial nephritis
Renal vasoconstriction
Reduced renal perfusion
Hyperkalemia
Sodium retention and edema
Gastrointestinal side effects Dyspepsia [69]
Heartburn [68]
Peptic ulcers
GI bleeding
Gastroduodenal ulcers
Cardiovascular side effects Myocardial infarction [60]
Thromboembolic events [68]
Atrial fibrillation
Heart stroke
Elevated blood pressure and Hardening of arteries [70]
hypertension Electrolyte imbalance
Hepatotoxic side effects Acute liver failure [71]
Others Urticaria, skin, and pulmonary [60]
infections
In contrast to the synthetic drugs that are grievous to human beings and the environ-
ment, herbal drugs are the symbol of safety as they are free from side effects, eco-
friendly, and locally available. Plants produce phytochemicals as secondary
metabolites to protect themselves from the both biotic and abiotic factors. Medicinal
plants with lead bioactive compounds play an important role in curing various dis-
eases [72]. Extraction of bioactive metabolites from plant material is the most
important step in testing its biological activity. When a whole extract is used,
chances are more for the synergistic effect between the active compounds in com-
parison to the isolation of each component [73]. The common anti-inflammatory
phytoconstituents found in herbal plants are steroids, flavonoids, alkaloids, poly-
phenols, glycosides, terpenoids, curcumins, Gamma-linolenic acid (GLA), linear
aliphatic alcohols, phenolic diterpenes, etc. Different mechanisms have been
explored for the anti-inflammatory potential of various herbal plants [74] (Fig. 4).
Plant secondary metabolites are produced by three biosynthetic pathways - shiki-
mate, acetate-malonate, and acetate-mevalonate pathway [75].
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1207
The enormous structural diversity of natural products (NP) and their medicinal sig-
nificance has led researchers to predict that screening natural resources will gener-
ate new lead compounds [76]. Although NSAIDs are extensively used as medication,
high doses can lead to GI, renal or liver abnormalities [8, 77] and other side effects
on human health whereas plant leads are with less toxicity or at safe levels [78].
Recurrence of symptoms is another major problem related to NSAIDs which falls
under synthetic drugs [79] but NPs are efficient and have a wide spectrum specific-
ity toward curing the disease [78]. Various research findings have proven the state-
ment [80, 81]. Thus, plant lead compounds are safe, efficacious, cost-effective, and
biocompatible alternatives to treat inflammatory diseases [82, 83] (Fig. 5).
Chemicals of plant origin can also serve as a basis for later pharmacological chemi-
cal modification to provide for drugs of improved specificity or potency compared
to NSAIDs [78].
Medicinal plants are a huge repository of biologically active compounds and can be
used to treat various diseases either as a crude material or as a pure compound [84].
1208 N. S. Nirmala et al.
Phyllanthus polyphyllus is a small shrub with one benzenoid and three aryl naph-
thalide lignans-based metabolites with medicinal importance. It inhibits the produc-
tion of NO and main proinflammatory cytokines such as TNFα, IL-12. It is useful in
the treatment of asthma, rheumatoid arthritis, and septic shock [84].
The polyphenolic extract of olive leaves inhibited COX-2 and suppressed the pro-
duction of a proinflammatory cytokine such as IL-6 and IL-1β [89]. The extra virgin
olive oil decreases the postprandial inflammatory response by reducing postprandial
lipopolysaccharide levels and diminishes the risk of heart diseases [86].
Carnosol and carnosic acids are two phenolic diterpenes extracted from S. officina-
lis and were reported as anti-inflammatory metabolites. Both the phenolic diter-
penes directly interacted with microsomal PGE [2] synthase (mPGES)-1 and
suppressed prostaglandin G2 (PGE2) production [91].
Fig. 6 Molecular mechanism of action of methanolic leaf extract of Bischofia javanica (MBJ)
toward anti-inflammatory effect
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1211
The ethanolic leaf extract of S. jollyanum was rich in alkaloids, tannins, saponins,
flavonoids, and cardiac glycoside content. Further the ethanolic extract S. jollyanum
was reported to reduce carrageenan-induced paw edema, egg albumin-induced paw
edema, and xylene-induced ear edema in experimental mice [94].
Roasted seeds, fruit infusion, fresh stem bark, and leaves of K. africana have been
reported to have potential therapeutic effect against inflammation [95].
Nowadays Quorum sensing is emerging as the potential target for treating bacterial
infection which causes inflammation. Synthetic Coumaperine derivatives from
piper nigrum have been reported to inhibit Quorum sensing and block the NF-κB
pathway, which ultimately leads to the inhibition of inflammation [96].
6.1 Alkaloids
6.2 Terpenoids
Terpenoids are one of the most important families of natural compounds known for
their medicinal value. Terpenoids are the most widespread class of secondary
metabolites found in plants and lower invertebrates. Terpenoids are derived from
mevalonate through a common biosynthetic pathway and are named terpenoids,
terpenes or isoprenoids [136]. Most of the terpenes isolated from plant sources are
well-known inhibitors of NF-κB signaling, the major regulator in the pathogenesis
of inflammatory diseases and thus possessing anti-inflammatory applications [137].
Terpenoids are having 1, 2, 3, 4, 5, 6, and 8 isoprenoid C5 residues and are clas-
sified into hemi, mono, sesqui, di, sester, tri, and tetra terpenoids (carotenoids)
respectively [138]. The exact molecular target of the terpenoids remains unknown
as in the case of most plant extracts. Pharmaceutical studies with monoterpenoids
indicate that they may have some therapeutic potential whereas few studies indicate
the direct role of monoterpenoids in the inhibition of NF-kB signaling.
Sesquiterpenoids are known to be present in several traditional natural remedies.
Though several thousands of sesquiterpene structures have been characterized, ses-
quiterpene lactones are the ones which are most frequently found in natural formu-
lations. Physiologically active diterpenoids include varied classes like aphidicolin,
gibberellins, phorbols, taxanes, etc. They do not directly affect the NF-κB system,
although they can have indirect effects. Triterpenoids are the major substituent in
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1213
Table 3 Summary of the various alkaloids reported from medicinal plants and their mode of anti-
inflammatory action
Class of Mode of anti-
alkaloid Lead compound(s) Plant species inflammatory action References
Indole Caulerpin Caulerpara Reduction of [102]
cemosa leukocyte cells in
number
Cassiaindoline Cassia alata Inhibition of [103]
anti-inflammatory
effect with edema
Strictosidine Uncaria Inhibition of NO [104]
rhynchophylla production
Geleganimine B Gelsemium Inhibition on the [105]
elegans Lipopolysaccharide-
stimulated NO
production
Picrinine Alstonia Suppression of 5-LOX [106]
Vallesamine scholaris Dual inhibition on
COX-2 and 5-LOX
Scholaricine Inhibition on COX-2
Alsmaphorazine A Alstonia Inhibit the production [107]
pneumatophora of NO
Mitraphylline Uncaria Reduction in levels of [108]
tomentosa proinflammatory
cytokines
Evodiamine Evodia efructus Reduction in levels of [109]
proinflammatory
mediators
Suppression of
phosphorylation of
IκBα
Inhibition activation of
NF-κB
Carbazole Clausine D Clausena Reduction in the [110]
lansium production of
superoxide anion
Clauemarazole E Clausena Inhibition of NO [111]
emarginata production
Murrayakonine A Murraya Inhibition of [112]
Mukolidine koenigii proinflammatory
O-methyl murrayamine A cytokines (IL-6 and
TNF-α)
Girinimbine Murraya Reduction in the [113]
koenigii levels of NO, PGE2,
TNF-α, and IL-6 due
to the suppression of
COX-2 enzyme and
iNOS
(continued)
1214 N. S. Nirmala et al.
Table 3 (continued)
Class of Mode of anti-
alkaloid Lead compound(s) Plant species inflammatory action References
β-Carboline 4-Methoxy-5- Picrasma Suppression in the [114]
hydroxycanthin-6-one quassioides production or [115]
expression of TNF-α,
NO, and iNOS
Dichotomide III, Stellaria Inhibition of [116]
dichotomide X, stellarine dichotoma and production of NO [117]
A, stellarine B, 1-acetyl-3- Neolitsea
methoxycarbonyl-b daibuensis
carboline, and
daibucarboline A
7-Methoxy-(9H-b- Eurycoma Reduction of [118]
carbolin-1-il)-1-propenoic longifolia production of NO,
(7-MCPA) PGE2, and IL-6, and
also suppression of
the expression of
iNOS and COX-2
Harmine Peganum Inhibition of NF-κB [119]
harmala L. transactivity
Quinoline Leucophyllidine Leuconotis Reduction in the [120]
eugenifolius production of NO and
the expression of
iNOS
Cryptolepine Cryptolepiss Inhibition of the NO [121]
anguinolenta production and the
level of NF-κB
Skimmianine Ruta graveolens Suppression of [122]
L. proinflammatory
cytokines production
and the antioxidant
effect
Evolitrine Acronychia Suppression of [123]
pedunculata proinflammatory
cytokines
Oleracimine Portulaca Inhibition of the [124]
oleracea expressions of COX-2
and iNOS
Antidesmone Ajuga Suppression of the [125]
decumbens excess production of
Thunb. proinflammatory
cytokines
Regulation of
MAPK- NFκB
signaling pathways
(continued)
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1215
Table 3 (continued)
Class of Mode of anti-
alkaloid Lead compound(s) Plant species inflammatory action References
Isoquinoline (+)-1-Hydroxy- Lycorisradiata Selective inhibition of [126]
ungeremine COX-2
(+)-N-methoxylcarbonyl-2-
dimethyl-isocorydione
Palmatine Coptis japonica Inhibition of the [127]
production and
expression of ROS,
NO, matrix
metalloproteinase-9
(MMP-9), and iNOS
Columbamine Tinospora Inhibition on [128]
sagittata LPS-induced NO
production and
TNF-α-induced
NF-κB activation
Glaucine Glaucium Reduction in the [129]
flavum Crantz production of TNF-α
and IL-6 and
enhanced the
production of IL-10
Pseudo Coptisine Corydalis Inhibition of the [130]
turtschaninovii activation of NF-κB
by the suppression of
ERK and p38
phosphorylation
Norisoboldine Lindera Reduction in [131]
aggregata infiltration of
(Sims.) Kosterm inflammatory cells
Erythraline Erythrina Suppression of NO [132]
crista-galli L. production
Chelidonine Chelidonium Suppression of the [133]
majus production of
proinflammatory
mediators
Warifteine Cissampelos Reduction in [134]
sympodialis neutrophil cell
adhesion and
migration
Delavatine A Incarvillea Suppression in the [135]
delavayi release of
proinflammatory
mediators by
inhibiting gene
expression
1216 N. S. Nirmala et al.
Table 4 Summary of terpenoids reported from medicinal plants and their mode of anti-
inflammatory action
Mechanism of
Class of the Lead anti-inflammatory
terpenoid compound(s) Plant species action References
Monoterpenoids Aucubin Aucuba japonica IκBα degradation [140]
Catalposide Catalpa ovata IκBα degradation [141]
Sesquiterpenoids Artemisolide Artemisia asiatica IKKβ inhibition on [142]
Cys-179
Nepalolide A Carpesium IκB phosphorylation [143]
nepalense
Diterpenoids Acanthoic Acanthopanax IκBα phosphorylation [144]
Acid Koreanum Nakai
Tanshinone Salvia IKKα /β and NIK [145]
IIA miltiorrhiza inhibition
Triterpenoids Avicins Acacia victoriae DNA binding [146, 147]
Betulinic acid Betula alba IKKα inhibition [148, 149]
Tetraterpenoids Lycopene Psidium guajava NF-κB translocation [150, 151]
β- carotene Momordica IκBα degradation, DNA [152, 153]
cochinchinensis binding
several Chinese herbal remedies, such as ginseng and Platycodon. Carotenoids are
powerful antioxidants which have therapeutic effects in several chronic illnesses,
such as cardiovascular disease and osteoporosis. In addition, they play major role
against inflammatory responses and cancer, suggesting their role in modulating
redox-sensitive signaling pathways, such as NF-κB signaling. Most terpenes are
known to have upstream targets common to both NF-κB signaling pathway and
MAPK pathway which ultimately lead to the suppression of inflammatory responses
[139]. Some of the examples of terpenoids showing potent anti-inflammatory poten-
tial have been listed (Table 4).
6.3 Polyphenols
7 Conclusion
Localization and elimination of harmful agents is the major aim of inflammation but
uncontrolled and chronic inflammatory responses lead to many inflammatory dis-
eases. The synthetic drugs such as NSAIDs widely used in the treatment of many
inflammatory diseases are having deleterious side effects in the gastrointestinal,
cardiovascular, and renal systems. Development of less toxic and more effective
anti-inflammatory agents is an ultimate but challenging task for a pharmaceutical
chemist. However, plant metabolites with diverse and potential lead compounds are
candidates for the design and discovery of new anti-inflammatory drugs. Application
of medicinal plants for the treatment of inflammatory diseases has been promising.
This chapter has provided a concise summary describing the potency of plant com-
pounds with anti-inflammatory activities and their possible molecular mechanisms
that would help researchers engaged in discovery of potential phytochemical lead
compounds with anti-inflammatory potential.
1218 N. S. Nirmala et al.
Table 5 Summary of the various polyphenols reported from medicinal plants and their mode of
anti-inflammatory action
Class of phenolic Mode of anti-
compound Lead compound(s) Plant species inflammatory action References
Stilbenes Resveratrol Polygonum Reduction in the [161]
cuspidatum expression of TNF-α,
IL-6, c-Jun N -
terminal kinase
(JNK-1), IκB Kinase β
(IKKβ), NF-κ B,
C-reactive protein
(CRP), suppressor of
cytokine signaling 3
(SOCS-3)
Vaccinium Inhibit NF-κB and [162]
macrocarpon JAK-STAT signaling
pathways
Piceatannol Passiflora Suppression of TNF-α, [163]
edulis NF-Κb, iNOS, IL-6, NO
Pterostilbene Vaccinium Inhibit the production [164]
corymbosum of TNF-α, IL-1 β,
IL-4, COX-2, NF- κB
Lignans Arctigenin Arctium lappa Inhibition of NO, [165]
iNOS, IL-6, TNF-α
Secoisolariciresinol Linumusita Inhibition of NF- κB [166]
diglucoside (SDG) tissimum signaling pathway
Phenolic acid Curcumin Curcuma longa Inhibition of [167]
proinflammatory
signaling cascades,
such as the NF𝜅B-,
MAPK, COX, and
LOX-pathways.
Downregulation of
cytokines like TNF𝛼,
IL-1𝛽, and IL-6.
Blocking the
expression of cell
adhesion molecules
(e.g., ICAM-1)
Hydroxycinnamic Caffeic acid Rhodiola sacra Reduction in the level [168]
acid phenethyl ester of TNF-α, IL-1β,
(CAPE) NF-κB
Caffeic acid Allium sativum Inhibition of NF-κB [169]
and AP-1
p-coumaric acid Cornifructus Inhibition of NF- κB [170]
signaling pathway
Rosmarinic acid Blechnum Reduction in the level [171]
brasiliense of TNF-α, IL-1β
Chlorogenic acid Cymbopogo Inhibition of NF-κB [172]
ncitratus signaling pathway
Ferulic acid Triticum Inhibition of TNF-α, [173]
aestivum IL-6, and NO
(continued)
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1219
Table 5 (continued)
Class of phenolic Mode of anti-
compound Lead compound(s) Plant species inflammatory action References
Hydroxybenzoic Gallic acid Camellia Inhibition of 5-LOX [174]
acid (HBA) sinensis activity
Protocatechuic acid Eucommia Reduce the level of [175]
(PCA) ulmoides IL-2, IL-6, TNF-α
Phenolic Hydroxytyrosol Olea europaea Inhibition of iNOS, [176]
derivatives COX-2, NF-κB,
STAT-1α, Interferon
regulatory factors
(IRF-1)
Oleocanthal Olea europaea Inhibition of NO, [177]
Macrophage
inflammatory protein -
1α(MIP-1α), IL-1β,
TNF-α, IL-6
6-Shogaol Zingiber Inhibit the release of [178]
officinale NO, iNOS,
prostaglandin E2
(PGE2), IL-1β, TNF-α,
COX-2. Block the
signaling pathways
such as NF-κB,
MAPK.
Oleuropein Olea europaea Downregulation of [179]
cyclooxygenase
(COX)-2, microsomal
prostaglandin E
synthase-1 (mPGES-
1), MAPK, NF-κB
signaling pathways,
IL-6, and TNF-α
cytokines
Magnolol Magnolia Suppression of NO, [180]
officinalis iNOS, PGE2, NF-κB
Tannins Prodelphinidin-B4-3′- Camellia Inhibition of COX-2, [181]
O-gallate (PDG) sinensis iNOS, PGE2, NO,
NF-κB
Penta-O-galloyl- β-D- Paeonia Inhibition of IL-8, [182]
glucose suffruticosa NF-κB, TNF-α
Paeonia Inhibition of iNOS, [183]
lactiflora COX-2
Gallotannin 15 Euphorbia Inhibition of NO, [184]
(1,2,3,4,6-penta-O- species iNOS, NF-κB
galloyl-β-D-glucose)
Gallotannin 23
(1,2,6-tri-O-galloyl-β-
D-allose)
(continued)
1220 N. S. Nirmala et al.
Table 5 (continued)
Class of phenolic Mode of anti-
compound Lead compound(s) Plant species inflammatory action References
Coumarins Osthole Cnidium Inhibit the production [185]
monnieri of NO, PGE2, TNF-α,
and IL-6. Suppressed
the activity of iNOS,
COX-2, NF-κB, and
p38 MAPK signaling
pathways
Scopoletin Canarium Inhibition of 5-LOX [186]
patentinervium activity
Daphnetin Daphne odora Inhibition of IL-1β, [187]
TNF-α, iNOS, and
COX-2, NO
6-isopentenyloxy-7- Gerbera Inhibition of NF-κB
methoxy coumarin serrata and signaling pathway
and Haplophyllum
8-isopentenyloxy7- pedicellatum
methoxy coumarin
Esculetin Fraxinus Suppression of [188]
rhynchophylla proinflammatory
cytokines (TNF-α,
IL-1β, IL-6) and
chemokine (CCL17)
through blocking of
NF-κB and STAT1
signaling pathway
Anthraquinones Chrysophanol Rheum Reduction in the levels [189]
palmatum of interleukin (IL)-4,
IL-5, and IL-13, tumor
necrosis factor
(TNF)-α, inducible
nitric oxide synthase,
and blocking of
NF-kB signaling
pathway
Emodin Aloe vera Suppression of IL-1β, [190]
IL-6, TNF-α, TLR4/
NF-κB signaling
pathway, vascular cell
adhesion molecule 1
(VCAM1), and
intercellular adhesion
molecule 1 (ICAM-1)
(continued)
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1221
Table 5 (continued)
Class of phenolic Mode of anti-
compound Lead compound(s) Plant species inflammatory action References
Flavonoids Homoplantaginin Salvia plebeia Suppression of TLR4 [191]
and NLRP3 signaling
Baicalin Scutellaria Inhibition of NLRP3 [192]
baicalensis inflammasome
Licoflavanone Glycyrrhiza Decreased [193]
glabra L. proinflammatory
cytokines
and COX-2/iNOS
expression levels
Apigenin Justicia Reduced the levels of [194]
gendarussa NF-κB, COX-2,
PGE2, IL-1β, and
TNF-α; elevated the
level of anti-
inflammatory
cytokine, IL-10
Kaempferol Portulaca Decreased the [195]
oleracea L. expression levels of
iNOS and COX-2
Barringoside I Barringtonia Inhibitory effects on [196]
racemosa LPS-induced NO
production
References
8. Dewanjee S, Dua TK, Sahu R (2013) Potential anti-inflammatory effect of Leea macro-
phylla Roxb. leaves: a wild edible plant. Food Chem Toxicology 59:514–520. https://doi.
org/10.1016/j.fct.2013.06.038
9. Divya T, Latha P, Usha K, Anuja G, Suja S, Shyamal S, Shine V, Sini S, Shikha P,
Rajasekharan S (2009) Anti-inflammatory, analgesic and anti-lipid peroxidative properties of
Wattakakavolubilis (Linn. f.) Stapf. Indian J Nat Prod Resour 8:137–141
10. Sofidiya MO, Imeh E, Ezeani C, Aigbe FR, Akindele AJ (2014) Antinociceptive and anti-
inflammatory activities of ethanolic extract of Alafia barteri. Rev Bras Farmacogn 24:348–354
11. Singh AG, Kumar A, Tewari DD (2012) An ethnobotanical survey of medicinal plants used in
Terai forest of western Nepal. J Ethnobiol Ethnomed 8(1):1–15
12. Ashutosh M, Kumar PD, Ranjan MM, Susil K, Ashutosh M (2009) Phytochemical screening
of Ichnocarpus Frutescens plant parts. Int J Pharmacogn Phytochem Res 1(1):5–7
13. Picchi V, Migliori C, Scalzo RL, Campanelli G, Ferrari V, Di Cesare LF (2012) Phytochemical
content in organic and conventionally grown Italian cauliflower. Food Chem 130(3):501–509
14. Dey PM (2012) Methods in plant biochemistry, Vol 1. Academic Press
15. McDowell PJ, Diver S, Yang F, Borg C, Busby J, Brown V, Grandison T (2021) The inflam-
matory profile of exacerbations in patients with severe refractory eosinophilic asthma receiv-
ing mepolizumab (the MEX study): a prospective observational study. Lancet Respir Med
9(10):1174–1184
16. Kaser A, Martínez-Naves E, Blumberg RS (2010) Endoplasmic reticulum stress: implications
for inflammatory bowel disease pathogenesis. Curr Opin Gastroenterol 26(4):318
17. Mirkov MU, Verstockt B, Cleynen I (2017) Genetics of inflammatory bowel disease: beyond
NOD2. Lancet Gastroenterol Hepatol 2(3):224–234
18. Ramos GP, Papadakis KA (2019) Mechanisms of disease: inflammatory bowel diseases. In:
Mayo Clinic proceedings, vol 94(1). Elsevier, pp 155–165
19. Guan Q (2019) A comprehensive review and update on the pathogenesis of inflammatory
bowel disease. J Immunol Res 1:7247238. https://doi.org/10.1155/2019/7247238
20. Grant RK, Jones GR, Plevris N, Lynch RW, Jenkinson PW, Lees CW, Arnott ID (2021)
The ACE (Albumin, CRP and Endoscopy) index in acute colitis: a simple clinical index on
admission that predicts outcome in patients with acute ulcerative colitis. Inflamm Bowel Dis
27(4):451–457
21. Langan RC, Gotsch PB, Krafczyk MA, Skillinge DD (2007) Ulcerative colitis: diagnosis and
treatment. Am Fam Physician 76(9):1323–1330
22. Ricanek P, Lunde LK, Frye SA, Støen M, Nygård S, Morth JP, Tønjum T (2015) Reduced
expression of aquaporins in human intestinal mucosa in early stage inflammatory bowel dis-
ease. Clin Exp Gastroenterol 8:49
23. Mekhjian HS, Switz DM, Watts HD, Deren JJ, Katon RM, Beman FM (1979) National
Cooperative Crohn’s disease study: factors determining recurrence of Crohn’s disease after
surgery. Gastroenterology 77(4):907–913
24. Mandal S, Barnett J, Brill SE, Brown JS, Denneny EK, Hare SS, Hurst JR (2021) ‘Long-
COVID’: a cross-sectional study of persisting symptoms, biomarker and imaging abnormali-
ties following hospitalisation for COVID-19. Thorax 76(4):396–398
25. Brinster NK (2008) Dermatopathology for the surgical pathologist: a pattern based approach
to the diagnosis of inflammatory skin disorders (part I). Adv Anat Pathol 15(2):76–96
26. Rabinowitz LG, Esterly NB (1993) Inflammatory bullous diseases in children. Dermatol Clin
11(3):565–581
27. Smits T, Robles CA, van Erp PE, van de Kerkhof PC, Gerritsen MJP (2005) Correlation
between macroscopic fluorescence and protoporphyrin IX content in psoriasis and actinic
keratosis following application of aminolevulinic acid. J Investig Dermatol 125(4):833–839
28. Streit WJ, Mrak RE, Griffin WST (2004) Microglia and neuroinflammation: a pathological
perspective. J Neuroinflammation 1(1):1–4
29. Ebert SE, Jensen P, Ozenne B, Armand S, Svarer C, Stenbaek DS, Pinborg LH (2019)
Molecular imaging of neuroinflammation in patients after mild traumatic brain injury: a lon-
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1223
gitudinal 123I-CLINDE single photon emission computed tomography study. Eur J Neurol
26(12):1426–1432
30. Mosley RL, Hutter-Saunders JA, Stone DK, Gendelman HE (2012) Inflammation and adap-
tive immunity in Parkinson’s disease. Cold Spring Harb Perspect Med 2(1):a009381
31. Mihaylova S, Killian A, Mayer K, Pullamsetti SS, Schermuly R, Rosengarten B (2012)
Effects of anti-inflammatory vagus nerve stimulation on the cerebral microcirculation in
endotoxinemic rats. J Neuroinflammation 9(1):1–11
32. Garden GA (2013) Epigenetics and the modulation of neuroinflammation. Neurotherapeutics
10(4):782–788
33. Streit WJ, Xue QS, Braak H, Del Tredici K (2014) Presence of severe neuroinflammation
does not intensify neurofibrillary degeneration in human brain. Glia 62(1):96–105
34. Mayer CL, Huber BR, Peskind E (2013) Traumatic brain injury, neuroinflammation, and
post-traumatic headaches. Headache 53(9):1523–1530
35. Wang Q, Liu Y, Zhou J (2015) Neuroinflammation in Parkinson’s disease and its potential as
therapeutic target. Transl Neurodegener 4(1):1–9
36. Meraz-Ríos MA, Toral-Rios D, Franco-Bocanegra D, Villeda-Hernández J, Campos-Peña V
(2013) Inflammatory process in Alzheimer’s disease. Front Integr Neurosci 7:59
37. Barnum CJ, Tansey MG (2012) Neuroinflammation and non-motor symptoms: the dark pas-
senger of Parkinson’s disease? Curr Neurol Neurosci Rep 12(4):350–358
38. Kösling S (2008) Encyclopedia of diagnostic imaging. Springer-Verlag, Berlin Heidelberg,
New York
39. Crispian S (2008) Oral and maxillofacial medicine: the basis of diagnosis and treatment, 2nd
edn. Churchill Livingstone, Edinburgh, p 356. ISBN 978-0443068188
40. Rogers K (ed) (2010) The digestive system, 1st edn. Britannica Educational Pub., in associa-
tion with Rosen Educational Services, New York, p 146. ISBN 978-1615301317
41. Stewart MG, Selesnick S (eds) (2010) “35”. Differential diagnosis in otolaryngology head
and neck surgery. Thieme, New York. ISBN 9781604062793
42. Baelum V, Lopez R (2004) Periodontal epidemiology: towards social science or molecular
biology? Community Dent Oral Epidemiol 32(4):239–249
43. Nicchio I, Cirelli T, Nepomuceno R et al (2021) Polymorphisms in genes of lipid metabolism
are associated with Type 2 diabetes mellitus and periodontitis, as comorbidities, and with the
subjects’ periodontal, glycemic, and lipid profiles. J Diabetes Res 2021:1049307. PMCID:
PMC8601849
44. Braun-Falco O (2000) Dermatology: with 281 tables, 2nd completely rev edn. Springer,
Berlin, p 1166. ISBN 9783540594529
45. Andrade-Oliveira V, Foresto-Neto O, Watanabe IKM, Zatz R, Câmara NOS (2019)
Inflammation in renal diseases: new and old players. Front Pharmacol 10:1192
46. Verma VK, Kumar A, Bhardwaj G, Kumar S, Kumar M, Chandan KK (2015) Anaesthesia for
chronic renal disease and renal transplant: an update. J Evol Med Dent Sci 4(19):3346–3365
47. Floege J (2013) Primary glomerulonephritis: a review of important recent discoveries. J
Kidney Res Clin Pract 32(3):103–110
48. Kitching AR (2002) Cytokines, T cells and proliferative glomerulonephritis. Nephrology
7(5):244–249
49. Lovisa S, LeBleu VS, Tampe B, Sugimoto H, Vadnagara K, Carstens JL, Kalluri R (2015)
Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in
renal fibrosis. Nat Med 21(9):998–1009
50. Caravaca-Fontán F, Shabaka A, Sánchez-Álamo B, de Lorenzo A, Díaz M, Blasco M,
Rodríguez E, Sierra-Carpio M, Malek Marín T, Urrestarazú A, Corona Cases C, Praga M,
Fernández-Juárez G (2020) Spanish Group for the Study of glomerular diseases (GLOSEN).
Recurrent acute interstitial nephritis: what lies beneath? Clin Kidney J 14(1):197–204.
https://doi.org/10.1093/ckj/sfaa018
51. Meyers CM (2014) “Chronic tubulointerstitial disease”, National Kidney Foundation primer on
kidney diseases. Elsevier, pp 390–396. https://doi.org/10.1016/b978-1-4557-4617-0.00045-5.
ISBN 9781455746170
1224 N. S. Nirmala et al.
72. Khumalo GP, Van Wyk BE, Feng Y, Cock IE (2022) A review of the traditional use of
Southern African medicinal plants for the treatment of inflammation and inflammatory pain.
J Ethnopharmacol 283:114436. https://doi.org/10.1016/j.jep.2021.114436
73. Azab A, Nassar A, Azab AN (2016) Anti-inflammatory activity of natural products. Molecules
21(10):13–21
74. Yatoo M, Gopalakrishnan A, Saxena A, Parray OR, Tufani NA, Chakraborty S, Iqbal H
(2018) Anti-inflammatory drugs and herbs with special emphasis on herbal medicines for
countering inflammatory diseases and disorders-a review. Recent Patents Inflamm Allergy
Drug Discov 12(1):39–58
75. Wills RB, Bone K, Morgan M (2000) Herbal products: active constituents, modes of action
and quality control. Nutr Res Rev 13(1):47–77
76. Khazir J, Mir BA, Mir SA, Cowan D (2013) Natural products as lead compounds in drug dis-
covery. J Asian Nat Prod Res 15(7):764–788. https://doi.org/10.1080/10286020.2013.798314
77. Qandil AM (2012) Prodrugs of nonsteroidal anti-inflammatory drugs (NSAIDs), more than
meets the eye: a critical review. Int J Mol Sci 13(12):17244–17274
78. Ahmad B, Friar EP, Vohra MS, Garrett MD, Serpell CJ, Fong IL, Wong EH (2020)
Mechanisms of action for the anti-obesogenic activities of phytochemicals. Phytochemistry
180:112513. https://doi.org/10.1016/j.phytochem.2020.112513
79. Jo WS, Yang KM, Choi YJ, Jeong CH, Ahn KJ, Nam BH, Lee SW, Seo SY, Jeong MH (2010)
In vitro and in vivo anti-inflammatory effects of pegmatite. Mol Cell Toxicol 6:195202.
https://doi.org/10.1007/s13273-010-0027-0
80. Gomez-Flores R, Calderon CL, Scheibel LW, Tamez-Guerra P, Rodriguez-Padilla C, Tamez-
Guerra R, Weber RJ (2000) Immunoenhancing properties of Plantago major leaf extract.
Phytother Res 14(8):617–622
81. Ray B, Chauhan NB, Lahiri DK (2011) The “aged garlic extract”: (AGE) and one of its
active ingredients S-allyl-L-cysteine (SAC) as potential preventive and therapeutic agents for
Alzheimer’s disease (AD). Curr Med Chem 18:330613
82. Anand U, Jacobo-Herrera N, Altemimi A, Lakhssassi N (2019) A comprehensive review on
medicinal plants as antimicrobial therapeutics: potential avenues of biocompatible drug dis-
covery. Meta 9(11):258
83. Uddin G, Rauf A, Siddiqui BS, Muhammad N, Khan A, Shah SUA, Rauf DA (2014)
Anti-nociceptive, anti-inflammatory and sedative activities of the extracts and chemical
constituents of Diospyros lotus L. Phytomedicine 21:954–959. https://doi.org/10.1016/j.
phymed.2014.03.001
84. Kumar S, Bajwa BS, Kuldeep S, Kalia AN (2013) Anti-inflammatory activity of herbal
plants: a review. Int J Adv Pharm Biol Chem 2(2):272–281
85. Wal P, Saraswat N, Pal RS, Wal A, Chaubey M (2019) A detailed insight of the anti-
inflammatory effects of curcumin with the assessment of parameters, sources of ROS and
associated mechanisms. Open Med J 6(1):64
86. Ghasemian M, Owlia S, Owlia MB (2016) Review of anti-inflammatory herbal medicines.
Adv Pharmacol Sci 2016:9130979
87. Raditic DM, Bartges JW (2014) The role of chondroprotectants, nutraceuticals, and nutrition
in rehabilitation. In: Canine rehabilitation and physical therapy. WB Saunders, pp 254–276
88. Lattanzio F, Greco E, Carretta D, Cervellati R, Govoni P, Speroni E (2011) In vivo anti-
inflammatory effect of Rosa canina L. extract. J Ethnopharmacol 137(1):880–885
89. Aissaoui Y, Boukhari Y, Mahi MA (2021) The anti-inflammatory effect of polyphenolics
olive leaf extracts Olea europaea L. Phytothérapie. https://doi.org/10.3166/phyto-2021-0251
90. Wiart, C. (2007). Ethnopharmacology of medicinal plants: Asia and the Pacific. Springer
Science & Business Media
91. Bauer J, Kuehnl S, Rollinger JM, Scherer O, Northoff H, Stuppner H, Koeberle A (2012)
Carnosol and carnosic acids from Salvia officinalis inhibit microsomal prostaglandin E2
synthase-1. J Pharmacol Exp Ther 342(1):169–176
1226 N. S. Nirmala et al.
92. Lee S, Ha J, Park J, Kang E, Jeon SH, Han SB, Cho S (2021) Antioxidant and anti-
inflammatory effects of Bischofia javanica (Blume) leaf methanol extracts through the regu-
lation of Nrf2 and TAK1. Antioxidants (Basel) 10(8):1295
93. Emon NU, Alam S, Rudra S, Al Haidar IK, Farhad M, Rana MEH, Ganguly A (2021)
Antipyretic activity of Caesalpinia digyna (Rottl.) leaves extract along with phytoconstit-
uent’s binding affinity to COX-1, COX-2, and mPGES-1 receptors: in vivo and in silico
approaches. Saudi J Biol Sci 28(9):5302–5309
94. Uka E, Ewere EG, Effiong GS (2021) Anti-inflammatory potential of ethanol leaf extract of
Sphenocentrum jollyanum in experimental mice. GSC Adv Res Rev 7(1):146–156
95. Nabatanzi AM, Nkadimeng S, Lall N, Kabasa JD, McGaw JL (2020) Ethnobotany, phyto-
chemistry and pharmacological activity of Kigelia africana (Lam.) Benth. (Bignoniaceae).
Plants 9(6):753
96. Kadosh Y, Muthuraman S, Yaniv K, Baruch Y, Gopas J, Kushmaro A, Kumar RS (2021)
Quorum sensing and NF-κB inhibition of synthetic coumaperine derivatives from piper
nigrum. Molecules 26(8):2293
97. Gupta A, Kumar R, Ganguly R, Singh AK, Rana HK, Pandey AK (2021) Antioxidant, anti-
inflammatory and hepatoprotective activities of Terminalia bellirica and its bioactive com-
ponent ellagic acid against diclofenac induced oxidative stress and hepatotoxicity. Toxicol
Rep 8:44–52
98. Taha HS, El Bahr MK, Seif ENM (2009) In vitro studies on Egyptian Catharanthus roseus
(L.) G.Don. IV: manipulation of some amino acids as precursors for enhanced of indole alka-
loids production in suspension cultures. Aust J Basic Appl Sci 3(4):3137–3144
99. Seigler DS (1998) Pyrrolizidine, quinolizidine, and indolizidine alkaloids. In: Plant second-
ary metabolism. Springer, Boston, pp 546–567
100. Gautam R, Jachak SM (2009) Recent developments in anti-inflammatory natural products.
Med Res Rev 29(5):767–820
101. Bai R, Yao C, Zhong Z, Ge J, Bai Z, Ye X, Xie Y (2021) Discovery of natural anti-inflammatory
alkaloids: potential leads for the drug discovery for the treatment of inflammation. Eur J Med
Chem 213:113–165
102. de Souza ET, de Lira DP, de Queiroz AC, da Silva DJC, de Aquino AB, Mella EAC, Lorenzo
VP, de Miranda GEC, de Araujo-Junior JX, de Oliveira Chaves MC, Barbosa-Filho JM, de
Athayde-Filho PF, de Oliveira Santos BV, Alexandre-Moreira MS (2009) The antinociceptive
and anti-inflammatory activities of caulerpin, a bisindole alkaloid isolated from seaweeds of
the genus Caulerpa. Mar Drugs 7:689–704
103. Villaseñor IM, Sanchez AC (2009) Cassiaindoline, a new analgesic and anti-inflammatory
alkaloid from Cassia alata. Z Naturforsch C J Biosci 64(5–6):335–338. https://doi.
org/10.1515/znc-2009-5-605
104. Ma B, Wu C, Yang J, Wang R, Kano Y, Yuan D (2009) Three new alkaloids from the leaves of
Uncaria rhynchophylla. Helv Chim Acta 92(8):1575–1585
105. Qu J, Fang L, Ren X, Liu Y, Yu S, Li L, Bao X, Zhang D, Li Y, Ma S (2013) Bisindole
alkaloids with neural anti-inflammatory activity from Gelsemium elegans. J Nat Prod
76(12):2203–2209
106. Shang J, Cai X, Feng T, Zhao Y, Wang J, Zhang L, Yan M, Luo X (2010) Pharmacological
evaluation of Alstonia scholaris: anti-inflammatory and analgesic effects. J Ethnopharmacol
129(2):174–181
107. Koyama K, Hirasawa Y, Nugroho A, Hosoya T, Hoe T, Chan K, Morita H (2010)
Alsmaphorazines A and B, novel indole alkaloids from Alstonia pneumatophora. Org Lett
12(18):4188–4191
108. Rojas-Duran R, González-Aspajo G, Ruiz-Martel C, Bourdy G, Doroteo-Ortega V, Alban-
Castillo J, Robert G, Auberger P, Deharo E (2012) Anti-inflammatory activity of Mitraphylline
isolated from Uncaria tomentosa bark. J Ethnopharmacol 143(3):801–804
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1227
109. Fan X, Zhu J, Sun Y, Luo L, Yan J, Yang X, Yu J, Tang W, Ma W, Liang H (2017) Evodiamine
inhibits Zymosan-induced inflammation in vitro and in vivo: inactivation of NF-κB by inhib-
iting IκBα phosphorylation. Inflammation 40(3):1012–1027
110. Shen D, Chan Y, Hwang T, Juang S, Huang S, Kuo P, Thang T, Lee E, Damu A, Wu T (2014)
Constituents of the roots of Clausena lansium and their potential anti-inflammatory activity.
J Nat Prod 77(5):1215–1223
111. Xia H, Ou Yang G, Li C, Yang J, Ma J, Zhang D, Li Y, Li L, Zhang D (2015) Clauemarazoles
A–G, seven carbazole alkaloids from the stems of Clausena emarginata. Fitoterapia 103:83–89
112. Nalli Y, Khajuria V, Gupta S, Arora P, Riyaz-Ul-Hassan S, Ahmed Z, Ali A (2018) Correction:
four new carbazole alkaloids from Murraya koenigii that display anti-inflammatory and anti-
microbial activities. Org Biomol Chem 16(11):1994. https://doi.org/10.1039/c8ob90030b
113. Mohan S, Hobani Y, Shaheen E, Abou-Elhamd A, Abdelhaleem A, Alhazmi H, Abdelwahab
S (2020) Girinimbine from curry leaves promotes gastro protection against ethanol induced
peptic ulcers and improves healing via regulation of anti-inflammatory and antioxidant mech-
anisms. Food Funct 11(4):3493–3505
114. Fan H, Qi D, Yang M, Fang H, Liu K, Zhao F (2013) In vitro and in vivo anti-inflammatory
effects of 4-methoxy-5- hydroxycanthin-6-one, a natural alkaloid from Picrasma quassioides.
Phytomedicine 20(3–4):319–323. https://doi.org/10.1016/j.phymed.2012.11.016
115. Liu JF, Shao M, Zhai DW, Liu K, Wu LJ (2009) Protective effect of 4-methoxy-5-
hydroxycanthin-6-one, a natural alkaloid, on dextran sulfate sodium-induced rat colitis.
Planta Med 75(2):142–145. https://doi.org/10.1055/s-0028-1088390
116. Chen YF, Kuo PC, Chan HH, Kuo IJ, Lin FW, Su CR, Yang ML, Li DT, Wu TS (2010)
β-carboline alkaloids from Stellaria dichotoma var. lanceolata and their anti-inflammatory
activity. J Nat Prod 73(12):1993–1998. https://doi.org/10.1021/np1003627
117. Wong SL, Chang HS, Wang GJ, Chiang MY, Huang HY, Chen CH, Tsai SC, Lin CH, Chen
IS (2011) Secondary metabolites from the roots of Neolitsea daibuensis and their anti-
inflammatory activity. J Nat Prod 74(12):2489–2496. https://doi.org/10.1021/np100874f
118. Ngoc PB, Pham TB, Nguyen HD, Tran TT, Chu HH, Chau VM, Lee JH, Nguyen TD (2016)
A new anti-inflammatory β-carboline alkaloid from the hairy-root cultures of Eurycoma lon-
gifolia. Nat Prod Res 30(12):1360–1365. https://doi.org/10.1080/14786419.2015.1056187
119. Liu X, Li M, Tan S, Wang C, Fan S, Huang C (2017) Harmine is an inflammatory inhibitor
through the suppression of NF-κB signaling. Biochem Biophys Res Commun 489(3):332–338.
https://doi.org/10.1016/j.bbrc.2017.05.126
120. Deguchi J, Shoji T, Nugroho AE, Hirasawa Y, Hosoya T, Shirota O, Awang K, Hadi AH,
Morita H (2010) Eucophylline, a tetracyclic Vinylquinoline alkaloid from Leuconotis eugeni-
folius. J Nat Prod 73(10):1727–1729. https://doi.org/10.1021/np100458b
121. Olajide OA, Ajayi AM, Wright CW (2009) Anti-inflammatory properties of cryptolepine.
Phytother Res 23(10):1421–1425. https://doi.org/10.1002/ptr.2794
122. Ratheesh M, Sindhu G, Helen A (2013) Anti-inflammatory effect of quinoline alkaloid
skimmianine isolated from Ruta graveolens L. Inflamm Res 62(4):367–376. https://doi.
org/10.1007/s00011-013-0588-1
123. Ratnayake W, Suresh TS, Abeysekera AM, Salim N, Chandrika UG (2019) Acute anti-
inflammatory and anti-nociceptive activities of crude extracts, alkaloid fraction and evo-
litrine from Acronychia pedunculata leaves. J Ethnopharmacol 238:111827. https://doi.
org/10.1016/j.jep.2019.111827
124. Li CY, Meng YH, Ying ZM, Xu N, Hao D, Gao MZ, Zhang WJ, Xu L, Gao YC, Ying XX
(2016) Three novel alkaloids from Portulaca oleracea L. and their anti-inflammatory effects.
J Agric Food Chem 64(29):5837–5844. https://doi.org/10.1021/acs.jafc.6b02673
125. Lu X, Pu Y, Kong W, Tang X, Zhou J, Gou H, Song X, Zhou H, Gao N, Shen J (2017)
Antidesmone, a unique tetrahydroquinoline alkaloid, prevents acute lung injury via regulat-
ing MAPK and NF-κB activities. Int Immunopharmacol 45:34–42
1228 N. S. Nirmala et al.
126. Liu ZM, Huang XY, Cui MR, Zhang XD, Chen Z, Yang BS, Zhao XK (2015) Amaryllidaceae
alkaloids from the bulbs of Lycoris radiata with cytotoxic and anti-inflammatory activities.
Fitoterapia 101:188–193. https://doi.org/10.1016/j.fitote.2015.01.003
127. Jeon S, Kwon K, Shin S, Lee S, Rhee S, Han S, Lee J, Kim H, Cheong J, Ryu J, Min B, Ko K,
Shin C (2009) Inhibitory effects of Coptis japonica alkaloids on the LPS-induced activation
of BV2 microglial cells. Biomol Ther 17(1):70–78
128. Liu X, Hu Z, Shi Q, Zeng H, Shen Y, Jin H, Zhang W (2010) Anti-inflammatory and anti-
nociceptive activities of compounds from Tinospora sagittata (Oliv.) Gagnep. Arch Pharm
Res 33(7):981–987. https://doi.org/10.1007/s12272-010-0702-7
129. Remichkova M, Dimitrova P, Philipov S, Ivanovska N (2009) Toll-like receptor-mediated
anti-inflammatory action of glaucine and oxoglaucine. Fitoterapia 80(7):411–414
130. Yun KJ, Shin JS, Choi JH, Back NI, Chung HG, Lee KT (2009) Quaternary alkaloid, pseudo-
coptisine isolated from tubers of corydalis turtschaninovi inhibits LPS-induced nitric oxide,
PGE (2), and pro-inflammatory cytokines production via the down-regulation of NF-kappaB
in RAW 264.7 murine macrophage cells. Int Immunopharmacol 9(11):1323–1331. https://
doi.org/10.1016/j.intimp.2009.08.001
131. Luo Y, Liu M, Xia Y, Dai Y, Chou G, Wang Z (2010) Therapeutic effect of norisoboldine, an
alkaloid isolated from radix Linderae, on collagen-induced arthritis in mice. Phytomedicine
17(10):726–731. https://doi.org/10.1016/j.phymed.2010.01.013
132. Ozawa M, Kawamata S, Etoh T, Hayashi M, Komiyama K, Kishida A, Kuroda C, Ohsaki
A (2010) Structures of new Erythrinan alkaloids and nitric oxide production inhibitors
from Erythrina crista-galli. Chem Pharm Bull 58(8):1119–1122. https://doi.org/10.1248/
cpb.58.1119
133. Liao W, He X, Yi Z, Xiang W, Ding Y (2018) Chelidonine suppresses LPS-induced pro-
duction of inflammatory mediators through the inhibitory of the TLR4/NF-κB signaling
pathway in RAW264.7 macrophages. Biomed Pharmacother 107:1151–1159. https://doi.
org/10.1016/j.biopha.2018.08.094
134. Lima TF, Rocha JD, Guimarães-Costa AB, Barbosa-Filho JM, Decoté-Ricardo D, Saraiva
EM, Arruda LB, Piuvezam MR, Peçanha LM (2014) Warifteine, an alkaloid purified from
Cissampelos sympodialis, inhibits neutrophil migration in vitro and in vivo. J Immunol Res
2014:752923. https://doi.org/10.1155/2014/752923
135. Xie Q, Wu GZ, Yang N, Shen YH, Tang J, Zhang WD (2018) Delavatine A, an unusual iso-
quinoline alkaloid exerts anti-inflammation on LPS-induced proinflammatory cytokines pro-
duction by suppressing NF-κB activation in BV-2 microglia. Biochem Biophys Res Commun
502(2):202–208. https://doi.org/10.1016/j.bbrc.2018.05.144
136. Bouvier F, Rahier A, Camara B (2005) Biogenesis, molecular regulation and function of plant
isoprenoids. Prog Lipid Res 44(6):357–429. https://doi.org/10.1016/j.plipres.2005.09.003
137. Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J (2008) Terpenoids: natu-
ral inhibitors of NF-kappaB signaling with anti-inflammatory and anticancer potential. Cell
Mol Life Sci 65(19):2979–2999. https://doi.org/10.1007/s00018-008-8103-5
138. de las Heras B, Hortelano S (2009) Molecular basis of the anti-inflammatory effects of terpenoids.
Inflamm Allergy Drug Targets 8(1):28–39. https://doi.org/10.2174/187152809787582534
139. Kawai T, Akira S (2007) Signaling to NF-kappaB by Toll-like receptors. Trends Mol Med
13(11):460–469. https://doi.org/10.1016/j.molmed.2007.09.002
140. Jeong HJ, Koo HN, Na HJ, Kim MS, Hong SH, Eom JW, Kim KS, Shin TY, Kim HM (2002)
Inhibition of TNF-a and IL-6 production by aucubin through blockade of NF-kB activation
in RBL-2H3 mast cells. Cytokine 18:252–259
141. Kim SW, Choi SC, Choi EY, Kim KS, Oh JM, Lee HJ, Oh HM, Kim S, Oh BS, Kimm KC,
Lee MH, Seo GS et al (2004) Catalposide, a compound isolated from catalpa ovata, attenu-
ates induction of intestinal epithelial proinflammatory gene expression and reduces the sever-
ity of trinitrobenzene sulfonic acid-induced colitis in mice. Inflamm Bowel Dis 10:564–572
142. Kim BH, Lee JY, Seo JH, Lee HY, Ryu SY, Ahn BW, Lee CK, Hwang BY, Han SB, Kim
Y (2007) Artemisolide is a typical inhibitor of IkB kinase b targeting cysteine-179 residue
Anti-inflammatory Potential of Lead Compounds and Their Derivatives from Medicinal… 1229
177. Scotece M, Gómez R, Conde J, Lopez V, Gómez-Reino JJ, Lago F, Gualillo O (2012) Further
evidence for the anti-inflammatory activity of oleocanthal: inhibition of MIP-1α and IL-6 in
J774 macrophages and in ATDC5 chondrocytes. Life Sci 91(23–24):1229–1235
178. Ha SK, Moon E, Ju MS, Kim DH, Ryu JH, Oh MS, Kim SY (2012) 6-Shogaol, a ginger prod-
uct, modulates neuroinflammation: a new approach to neuroprotection. Neuropharmacology
63(2):211–223
179. Castejón ML, Rosillo MÁ, Montoya T, González-Benjumea A, Fernández-Bolaños JM,
Alarcón-de-la-Lastra C (2017) Oleuropein down-regulated IL-1β-induced inflammation and
oxidative stress in human synovial fibroblast cell line SW982. Food Funct 8(5):1890–1898
180. Lu SH, Hsu WL, Chen TH, Chou TC (2015) Activation of Nrf2/HO-1 signaling path-
way involves the anti-inflammatory activity of magnolol in Porphyromonas gingivalis
lipopolysaccharide- stimulated mouse RAW 264.7 macrophages. Int Immunopharmacol
29(2):770–778
181. Hou DX, Luo D, Tanigawa S, Hashimoto F, Uto T, Masuzaki S, Sakata Y (2007) Prodelphinidin
B-4 3′-O-gallate, a tea polyphenol, is involved in the inhibition of COX-2 and iNOS via the
downregulation of TAK1-NF-κB pathway. Biochem Pharmacol 74(5):742–751
182. Oh GS, Pae HO, Cho BM, Lee HS, Kim IK, Yun YG, Chung HT (2004) Penta-O-galloyl-
beta-D-glucose inhibits phorbol myristate acetate-induced interleukin-8 [correction of
intereukin-8] gene expression in human monocytic U937 cells through its inactivation of
nuclear factor-kappaB. Int Immunopharmacol 4(3):377–386
183. Lee SJ, Lee IS, Mar W (2003) Inhibition of inducible nitric oxide synthase and cyclooxygen-
ase-2 activity by 1, 2, 3, 4, 6-penta-O-galloyl-β-D-glucose in murine macrophage cells. Arch
Pharm Res 26(10):832–839
184. Kim MS, Park SB, Suk K, Kim IK, Kim SY, Kim JA, Kim SH (2009) Gallotannin isolated
from euphorbia species, 1, 2, 6-tri-O-galloyl-β-D-allose, decreases nitric oxide production
through inhibition of nuclear factor-κ> B and downstream inducible nitric oxide synthase
expression in macrophages. Biol Pharm Bull 32(6):1053–1056
185. Fan H, Gao Z, Ji K, Li X, Wu J, Liu Y, Zhao F (2019) The in vitro and in vivo anti-inflammatory
effect of osthole, the major natural coumarin from Cnidium monnieri (L.) Cuss, via the
blocking of the activation of the NF-κB and MAPK/p38 pathways. Phytomedicine 58:152864
186. Mogana R, Teng-Jin K, Wiart C (2013) Anti-inflammatory, anticholinesterase, and anti-
oxidant potential of scopoletin isolated from Canarium patentinervium Miq.(Burseraceae
Kunth). Evid Based Complement Alternat Med 2013:734824
187. Das S, Mandal S (2018) Current developments on anti-inflammatory natural medicines.
Asian J Pharm Clin Res 11(8):61
188. Jeong NH, Yang EJ, Jin M, Lee JY, Choi YA, Park PH, Kim SH (2018) Esculetin from
Fraxinus rhynchophylla attenuates atopic skin inflammation by inhibiting the expression of
inflammatory cytokines. Int Immunopharmacol 59:209–216
189. Song G, Zhang Y, Yu S, Lv W, Guan Z, Sun M, Wang J (2019) Chrysophanol attenuates
airway inflammation and remodeling through nuclear factor-kappa B signaling pathway in
asthma. Phytother Res 33(10):2702–2713
190. Chen Y, Feng B, Yuan Y, Hu J, Zhao W, Jiang H, Du Z (2020) Aloe emodin reduces car-
diac inflammation induced by a high-fat diet through the TLR4 signaling pathway. Mediat
Inflamm 2020:6318520. https://doi.org/10.1155/2020/6318520
191. He B, Zhang B, Wu F, Wang L, Shi X, Qin W, Lin Y, Ma S, Liang J (2016) Homoplantaginin
inhibits palmitic acid-induced endothelial cells inflammation by suppressing TLR4 and
NLRP3 inflammasome. J Cardiovasc Pharmacol 67(1):93–101. https://doi.org/10.1097/
FJC.0000000000000318
192. Liu X, Liu C (2017) Baicalin ameliorates chronic unpredictable mild stress-induced depres-
sive behavior: involving the inhibition of NLRP3 inflammasome activation in rat prefrontal
cortex. Int Immunopharmacol 48:30–34. https://doi.org/10.1016/j.intimp.2017.04.019
193. Frattaruolo L, Carullo G, Brindisi M, Mazzotta S, Bellissimo L, Rago V, Curcio R, Dolce
V, Aiello F, Cappello AR (2019) Antioxidant and anti-inflammatory activities of flavanones
1232 N. S. Nirmala et al.
Tanima Bhattacharya, Debashrita Das, Hitesh Chopra, and Atif Amin Baig
1 Introduction
The term Alzheimer’s disease is named after Dr. Alois Alzheimer in 1906.
Alzheimer’s disease is a serious ailment of the brain that gradually progresses and
cannot be reversed back, slowly destroying thinking and memorizing skills. As a
result, the individual suffering from this disease finds it very difficult to perform
even the simplest of tasks. The symptoms of the disease usually vary from person to
person, but generally, they include having problems in language, unpredictable
behavior, and memory loss [1]. Individuals in the age group of the mid-60 s are most
likely to face the symptoms of Alzheimer’s disease. The majority of the elderly in
the clinic have evidence of neurofibrillary and vascular pathology [2]. Elderly
Tanima Bhattacharya and Debashrita Das contributed equally with all other contributors.
T. Bhattacharya
Department of Food and Nutrition, College of Human Ecology, Kyung Hee University,
Dongdaemun-gu Seoul, Republic of Korea
Nondestructive Bio-Sensing Laboratory, Department of Biosystems Machinery Engineering,
College of Agriculture and Life Science, Chungnam National University, Yuseong-Gu
Daejeon, Republic of Korea
D. Das
PRCS Technologies, Kolkata, West Bengal, India
H. Chopra
Chitkara College of Pharmacy, Chitkara University, Rajpura, Patiala, Punjab, India
A. A. Baig (*)
University Institute of Public Health, Faculty of Allied Health Sciences, The University of
Lahore, Lahore, Punjab, Pakistan
Institute of Molecular Biology & Biotechnology, The University of Lahore,
Lahore, Punjab, Pakistan
people having mild cognitive impairment are more prone to develop Alzheimer’s
disease. The prevalence of this disease globally is supposed to be as much as 24
million of which in the USA, the number of affected people are about 5.5 million.
In western societies, Alzheimer’s disease frequently causes dementia [3]. The grad-
ual loss in cognitive functioning, which includes remembering capacity, thinking
ability, and reasoning capacity, and loss of behavioral capabilities, which causes
hindrance to proper carrying out of the day-to-day activities, is known as dementia.
However, the rate at which Alzheimer’s disease will progress is variable and approx-
imately individuals suffering from Alzheimer’s disease survive for 3–11 years after
being diagnosed with the disease and some can even survive for as much as 20 years.
During the time of diagnosis of the disease, the extent of impairment can eventually
directly impact the life expectancy of the affected individual. The leading cause
behind the disease is the development of proteins like amyloid and tau abnormally
inside and around the cells of the brain, forming plaques and tangles within the cells.
Figure 1 represents the pathophysiology of Alzheimer’s disease with amyloid
precursor protein causing cognitive dysfunction in human beings [4]. Considering
the alarmingly increasing mortality rate from Alzheimer’s disease, it is essential to
consider the natural foods that can act as medicines and help prevent and treat the
occurrence of Alzheimer’s disease successfully [5]. According to current trends,
several researches are being done for foods helping properly treat the disease. Thus,
different studies related to this subject are critically evaluated and reviewed so that
a better view of the recent progression in this subject area can be obtained.
Proper foods and nutritional support, including vitamins and curcumin, have a
prominent role in the survival of individuals suffering from Alzheimer’s disease.
Many studies have established a strong connection between nutrients and
Alzheimer’s disease, from which it can be known that the Mediterranean diet, vita-
mins, and curcumin are the three major role players in this regard. Swaminathan and
Jicha [6] suggest that approaching Alzheimer’s disease with proper nutrition can be
helpful in prevention, slowing down, or halting the usual progression of the occur-
rence of the disease.
Figure 2 represents how phyto-based drugs help prevent and treat the symptoms
and issues related to Alzheimer’s disease. It has been mentioned that plant flavo-
noids, vitamins, omega-3 fatty acids, metabolic substrates, antioxidants, and trace
minerals like dietary constituents can influence the cellular processes and pathways
connected with the neurodegeneration case of Alzheimer’s disease. In the following
section, certain specific foods acting as medicines in treating Alzheimer’s disease
will be extensively discussed, focusing on the recent research on this subject area.
Fig. 2 Mode of action of phyto-based drugs for the treatment of Alzheimer’s disease
1236 T. Bhattacharya et al.
Berries As per the opinion of Singh [7], berries containing plenty of nutritive and
bioactive substances have a vital role in preventing and providing therapeutic effects
for different neurodegenerative diseases like Alzheimer’s and Parkinson’s disease.
Berries, which include strawberry, grapes, blackberry, and blueberry, contain poly-
phenols that act as potent antioxidants as well as vitamins like B complex, A, C, and
E, which can help in delaying the occurrence of neurodegeneration along with
bringing about considerable improvement in the memory as well as cognitive activi-
ties of the frontal lobes present in the brain. The phytonutrients present in the berries
associate with the distinct signaling pathways, causing the folding of the proteins
and preventing neuroinflammation. Agarwal et al. [8] conducted a study in as many
as 925 patients aged between 58 and 98 years, and the connection between con-
sumption of strawberry and occurrence of Alzheimer’s-related dementia was evalu-
ated with the help of proportional hazard models. The study results found that
subjects who consumed a more significant number of strawberries experienced a
lower risk of dementia from Alzheimer’s. Vitamin C, anthocyanidins, flavonoids,
and pelargonidin were connected to lowering the risk of Alzheimer’s dementia.
Nuts Gorji et al. [9] conducted a study on the impact of nuts, especially almond,
hazelnut, and walnut, on acting against atrophy of brain and memory loss. The study
discussed the bioactive compounds present in these nuts and evaluated them as
effective supplements and natural foods acting as medicines in the treatment of
patients suffering from Alzheimer’s disease. It has also been found that these nuts
can essentially provide micro- and macronutrients along with essential phytochemi-
cals that can impact various pathways involved in the pathogenesis of Alzheimer’s
disease, including oxidative stress, tau phosphorylation, amyloidogenesis, and neu-
rogenesis. Similarly, in the literature of Chauhan and Chauhan [10], it has been
mentioned that walnuts contain different components showing antioxidant and anti-
inflammatory features, which can help in the therapeutic treatment of Alzheimer’s
disease. Supplementation in the diet with walnuts can be essentially beneficial for
the improvement in cognitive functions as well as reducing the chances of faster
progression of Alzheimer’s disease.
Omega-3 Fatty Acids It has been experimentally proven that the intake of a diet
enriched in docosahexaenoic acid (DHA) can protect brain cells from cognitive
damage and neurodegenerative diseases like Alzheimer’s. Yet, there are signs from
other epidemiological studies suggesting an unpredictable relationship between
having omega-3 fatty acids in the diet and decreased risk of Alzheimer’s. The study
of Quinn et al. [11] found that the omega-3 fatty acids with shorter chains are more
effective than those with long chains in treating Alzheimer’s disease. On the other
hand, in Avallone et al.’s [12] study, omega-3 fatty acids have shown strong poten-
tial in having anti-inflammatory and metabolic features in managing neurodegen-
erative diseases like Alzheimer’s and Parkinson’s. It was found that treating such
patients with omega-3 fatty acids was safe and well tolerated when supplementation
is done at the early stages of the disorder.
Alzheimer’s Disease Treatment Using Natural Foods: A Overview 1237
Spices In Mirmosayyeb et al.’s [14] article, the role of common spices like that of
pepper, cinnamon, turmeric, saffron, and zingiber toward prevention or halting of
neurodegenerative disease like Alzheimer’s has been discussed, which acts by sup-
pressing the inflammatory pathways involved in the disease and by exhibiting the
antioxidant activity and inhibition of acetylcholinesterase and aggregation of
amyloid-β. Mohanty (2017) studied the importance and function of Indian spices in
treating Alzheimer’s disease and evidenced that the phytochemicals present in
Indian spices can be a potential therapeutic agent for treating memory loss and
dementia related to Alzheimer’s disease. However, more studies are needed to help
in isolation, identification, and characterization of the bioactive compounds present
in the spices, which can help in the inhibition of cholinesterase activities and thereby
preventing the occurrence of Alzheimer’s [15].
Seeds The polyphenolic compounds present in grape seeds help in the prevention
and treatment of Alzheimer’s disease, and it is also safe, tolerable, bioactive, and
bioavailable. The polyphenolic compounds also help in interfering with the patho-
genesis of Alzheimer’s disease [16]. Biswas et al.’s [17] study assessed the cholin-
esterase inhibition potential of the extracts from the seed of Tamarindus indica, and
the results showed that the seed extracts have the moderate potential of inhibiting
acetylcholinesterase. However, it also suggested that further studies are needed to
identify molecules that may act as potent cholinesterase inhibitors in treating
Alzheimer’s disease.
2 Phyto-Based Drugs
There are five drugs approved for clinical use for the treatment of Alzheimer’s dis-
ease based on their properties of inhibiting cholinesterase, including tacrine, done-
pezil, and galantamine. But these drugs have been shown to have limited effectiveness
and are associated with other side effects to the body [18]. For this reason, the
1238 T. Bhattacharya et al.
Table 1 List of plants having medicinal value and their mode of action for treating
Alzheimer’s disease
Name of the Useful portion ofMode of action against Alzheimer’s
medicinal plant the plant disease References
Convolvulus Convolamine and Reduces biomarkers of Alzheimer’s Bihaqi et al.
pluricaulis convolvine disease in male Wister rats (2012) [21]
Glycyrrhiza Glycyrrhizin Relieves the inflammation of neurons Song et al.
glabra and memory deficiency as prompted by (2013) [22]
systemic treatment with
lipopolysaccharide in mice
Withania Withanamides Cleaves off beta-site amyloid precursor Mahrous et al.
somnifera and withanolides protein cleaving enzyme 1 (BACE1) or (2017) [23]
β-site amyloid precursor protein-cleaving
enzyme and acetylcholinesterase and
inhibits them
Nardostachys Valeranone and Improves memory and learning Karkada et al.
jatamansi nardosinone processes in mice (2012) [24]
Ficus carica Quercetin Causes induction of bioactivity of Khojah and
neurons and fights against Alzheimer’s Edrada-Ebel
disease related to oxidative stress (2017) [25]
Coriandrum Coumarin Proliferates glial cells, Liu et al.
sativum Suppresses Aβ42, and activates (2016) [26]
extracellular kinase regulated by signals
researchers are trying to find an alternative drug source that can be safe and effective
for the treatment of the disease. It has been seen in the literature of Ayaz et al. [19]
that curcumin can modulate inflammatory pathways involved in the central nervous
system, inhibit the aggregation of amyloid-β, as well as decrease the load of free
radicals in the body. For this reason, curcumin-based drugs can be essentially help-
ful to prevent or treat Alzheimer’s disease. Certain flavonoids like catechins and
myricetin help inhibit aggregation of amyloid-β and act as antioxidants, making
them a potent phyto-based drug for treating Alzheimer’s disease. Similarly, in the
study of Ovais et al. [20], certain important medicinal plants that have proper bioac-
tive substances and potentially act against Alzheimer’s disease have been exten-
sively highlighted. Among them, a few plants with their therapeutic properties
against Alzheimer’s have been discussed in Table 1.
Phyto-based drugs are essentially effective in treating Alzheimer’s disease and its
related symptoms. The study of Cacabelos et al. [27] suggests that plenty of phyto-
medicines exhibit promising outcomes in the therapeutic treatment of Alzheimer’s
disease. Along with customized nanomedicines, these phyto-based drugs could per-
form more effectively by efficiently delivering the bioactive compounds of the
Alzheimer’s Disease Treatment Using Natural Foods: A Overview 1239
medicines to the target sites in the body. Again, Girek and Szymański [18] discussed
the efficacy of tacrine that acts as the inhibitor of acetylcholinesterase, but it was
seen that there are several side effects of the drug. In this study, it was seen that the
side effects could be diminished if it is phyto-tacrine hybrid. It has also been high-
lighted that the phyto hybrids can exhibit anti-inflammatory and neuroprotective
effects, which certainly confirms the better efficacy of the phyto-based tacrine for
treating Alzheimer’s disease. D’Onofrio et al. [28] studied the role of phytochemi-
cals in the treatment of Alzheimer’s disease and concluded that the phytochemicals
can slow down the onset of Alzheimer’s disease by exhibiting anti-inflammatory,
anticholinergic, and antioxidant properties. In Karimi’s [29] manuscript, herbs like
Curcuma longa, Cyperus rotundus, and Thymus vulgaris have been shown to have
effective potential in the treatment of Alzheimer’s and these herbs could be later
produced as effective anti-Alzheimer’s drugs in the future. On the other hand, in the
manuscript of Ahmed and Gilani [30], the therapeutic potential of curcuminoid
mixture is believed to be more effective than that of curcumin alone in the treatment
of Alzheimer’s disease. From this, it can be stated that curcuminoids can be effec-
tively used for developing drugs derived from plant sources.
Figure 3 represents the various neuroprotective potential of different herbs and
their efficacy in performing antioxidant and anti-inflammatory functions in the body
[31]. The role of phytochemicals derived from various sources in treating
Alzheimer’s disease is described in Table 2.
Table 2 List of phytochemicals derived from different sources playing a beneficial role in the
treatment of Alzheimer’s disease
Name of Derived Pharmacological
the plant Phytochemical from impact Benefits References
Curcuma Curcumin Phenol Activation of Protects Nam et al.
longa regulation of cyclic neuroinflammation (2014)
adenosine neuritogenesis and [32];
monophosphate exhibits Hoppe
(AMP) response antioxidant et al.
element binding features (2013)
mediated by protein [33]
kinase C/extracellular
receptor kinase
Galanthus Galantamine Alkaloid Inhibition of Anti-inflammation, Furukawa
Sp. acetylcholinesterase, antioxidant, and et al.
accumulation of neuroprotection (2014)
microglial cells, and [34]
enhancement of
antioxidant enzymes
Abies Ligraminol Terpenoid Inhibition of the Protects against Xia et al.
holophylla E4-O-β-d- production of nitric neurodegenerative (2012)
xyloside oxide and [35]
neuroinflammatory
diseases
Citrus Naringenin Phenol Activation of Neuroprotection, Raza et al.
paradisi; signaling of nuclear anti-inflammatory (2013)
citrus factor erythroid effects, and [36]; Lou
sinensis 2-related factor/ antioxidant et al.
antioxidant response activities (2014)
element, upregulation [37]
of the antioxidant
enzymes, reduction
of nitric oxide, and
signaling of
cytokines
Morus Quercetin Phenol Scavenges free Antioxidant Pany et al.
alba radical, and inhibits activities and (2014)
cyclooxygenase-2 anti-inflammatory [38]
and nuclear factor properties
kappa light chain
enhancer of activated
B-cells
In the review by Howes et al. [39], the efficacy of Chinese herbal medicines
derived from plants like Polygala tenuifolia for treating Alzheimer’s disease has
been discussed. All the scientific evidences regarding the herbal medicines used
traditionally for the effective treatment of Alzheimer’s disease help in the under-
standing that phyto-based drugs can essentially be used for relieving the symptoms
and exhibit pharmacological properties pertinent to Alzheimer’s disease.
Alzheimer’s Disease Treatment Using Natural Foods: A Overview 1241
4 Toxicity Issues
Although it is evident from the above discussion that the phyto-based drugs have a
strong potential to treat Alzheimer’s disease, it is also of utmost importance to have
an in-depth understanding of whether there is any kind of toxicity-related issues or
not. This is why Mehla et al. [40] assessed the efficacy of Indian medicinal herbs
and, at the same time, evaluated the toxicity of the phyto drugs. The extract of
Centella asiatica, which can help in supporting enhanced working memory and bet-
ter self-related mood, is found to have good tolerance in various studies [41]. No
such toxicity issues have been reported for this herb up to a value of 1 g/kg of dos-
age when given orally. On the other hand, Bacopa monnieri extract was evaluated to
be safe and tolerable in healthy male helpers when given single and multiple doses
daily for 4 weeks.
Similarly, Sanka et al. [42] studied a shift in the paradigm of using drugs to treat
Alzheimer’s disease owing to fatal side effects toward the usage of phyto-based
herbal medicines. It is reported that herbal medicines exhibit a variety of benefits
over that of other drugs by having a significantly less adverse impact on the body.
This is because, unlike other medications, even if there is slight overdosage of the
phyto-based medicine, it would not be a vital issue for the body. Thus, it makes the
fact well evident that no such studies are reporting the toxicity of the phyto-based
drugs for the treatment of Alzheimer’s disease. Koynova and Tenchov [43] summa-
rized the efficacy of the extracts derived from plant sources and the drugs derived
from them for the purpose of treatment and also prevention of Alzheimer’s disease.
It stated that the phyto-based drugs are nowadays very well accepted globally and
that multifactorial type of pathogenesis of Alzheimer’s disease needs drugs cover-
ing a wide range of activities. For this reason, as phyto-based drugs possess a vari-
ety of compounds performing many mechanisms, these are estimated to be
non-harmful and more effective than drugs having a simple and single target for
treating complex disorders like Alzheimer’s disease.
It is highly evident from the overall discussion that Alzheimer’s disease has com-
plex pathophysiology and the rapid rise in the Alzheimer’s cases demands the effec-
tive treatment of the disease. Although the disease has been studied widely, as
reported by Opare Asamoah Botchway [44], tacrine was the first drug approved to
be used by the US Food and Drug Administration, mainly targeting the memory and
thinking-related signs of the disease. But still, it was seen that the drug had specific
side effects that were limited to the dosage, which involved anxiety, dizziness,
insomnia, nausea, and diarrhea, being typical for the cholinergic stimulation. Also,
owing to its potential to damage the liver, this drug was discontinued, and, later on,
1242 T. Bhattacharya et al.
6 Conclusion
The aging population of the developed and developing countries is increasing rap-
idly and therefore the frequency of occurrence of Alzheimer’s disease is likely to
almost double in every 20 years till the year 2040. Alzheimer’s disease will become
a public health burden in the upcoming years and be expensive. That is why it is
essential nowadays to focus on preventing and treatment of the Alzheimer’s disease
and not merely on the temporal relieving of the symptoms. Many studies suggest
that phyto-based drugs have a promising potential in curing Alzheimer’s disease
and slowing down the progression of its signs and symptoms.
Conflict of Interest The authors declare that there are no conflicts of interest, financial or otherwise.
References
1. Alzheimer’s Association (2016) 2016 alzheimer’s disease facts and figures includes a spe-
cial report on the personal financial impact of Alzheimer's on families. Alzheimer’s and
Dementia 12:459
2. Chertkow H, Feldman HH, Jacova C, Massoud F (2013) Definitions of dementia and pre-
dementia states in Alzheimer's disease and vascular cognitive impairment: consensus from
the Canadian conference on diagnosis of dementia. Alzheimers Res Ther 5:S2. https://doi.
org/10.1186/alzrt198
3. Mayeux R, Stern Y (2012) Epidemiology of Alzheimer disease. Cold Spring Harb Perspect
Med 2:a006239. https://doi.org/10.1101/cshperspect.a006239
4. Takahashi RH, Nagao T, Gouras GK (2017) Plaque formation and the intraneuronal accumula-
tion of β-amyloid in Alzheimer’s disease. Pathol Int 67:185. https://doi.org/10.1111/pin.12520
5. Botchway BOA, Moore MK, Akinleye FO, Iyer IC, Fang M (2018) Nutrition: review on the
possible treatment for Alzheimer’s disease. J Alzheimers Dis 61:867. https://doi.org/10.3233/
JAD-170874
6. Swaminathan A, Jicha GA (2014) Nutrition and prevention of Alzheimer’s dementia. Front
Aging Neurosci 6:282. https://doi.org/10.3389/fnagi.2014.00282
7. Singh R (2018) Current Alzheimers management with berries fruits therapy. J Public Health
Nutr 1:17. https://doi.org/10.35841/public-health-nutrition.1.2.17-24
8. Agarwal P, Holland TM, Wang Y, Bennett DA, Morris MC (2019) Association of strawber-
ries and anthocyanidin intake with alzheimer's dementia risk. Nutrients 11:3060. https://doi.
org/10.3390/nu11123060
9. Gorji N, Moeini R, Memariani Z (2018) Almond, hazelnut and walnut, three nuts for neuropro-
tection in Alzheimer's disease: a neuropharmacological review of their bioactive constituents.
Pharmacol Res 129:115–127. https://doi.org/10.1016/j.phrs.2017.12.003
10. Chauhan A, Chauhan V (2020) Beneficial effects of walnuts on cognition and brain health.
Nutrients. https://doi.org/10.3390/nu12020550
11. Quinn JF et al (2010) docosahexaenoic acid supplementation and cognitive decline in
Alzheimer disease: a randomized trial. JAMA J Am Med Assoc. https://doi.org/10.1001/
jama.2010.1510
12. Avallone R, Vitale G, Bertolotti M (2019) Omega-3 fatty acids and neurodegenerative dis-
eases: new evidence in clinical trials. Int J Mol Sci. https://doi.org/10.3390/ijms20174256
13. Manchali S, Chidambara Murthy KN, Patil BS (2012) Crucial facts about health benefits of
popular cruciferous vegetables. J Funct Foods. https://doi.org/10.1016/j.jff.2011.08.004
14. Mirmosayyeb O et al (2017) possible role of common spices as a preventive and therapeutic
agent for Alzheimer's disease. Int J Prev Med. https://doi.org/10.4103/2008-7802.199640
15. Singhal A, Bangar O, Naithani V (2012) Medicinal plants with a potential to treat
Alzheimer and associated symptoms. Int J Nutr Pharmacol Neurol Dis Ther. https://doi.
org/10.4103/2231-0738.95927
16. Pasinetti G (2010) Role of grape seed polyphenols in Alzheimer’s disease neuropathology.
Nutr Diet Suppl. https://doi.org/10.2147/nds.s6898
17. Biswas K et al (2017) Assessment of in-vitro cholinesterase inhibitory and thrombolytic poten-
tial of bark and seed extracts of Tamarindus indica (L.) relevant to the treatment of Alzheimer's
disease and clotting disorders. J Intercult Ethnopharmacol. https://doi.org/10.5455/
jice.20161229055750
18. Girek M, Szymański P (2019) Phyto-Tacrine hybrids as promising drugs to treat Alzheimer's
disease. Chemistry Select. https://doi.org/10.1002/slct.201803672
19. Ayaz M, Ullah F, Sadiq A, Kim MO, Ali T (2019) Editorial: natural products-based drugs:
potential therapeutics against Alzheimer's disease and other neurological disorders. Front
Pharmacol. https://doi.org/10.3389/fphar.2019.01417
1244 T. Bhattacharya et al.
40. Mehla J, Gupta P, Pahuja M, Diwan D, Diksha D (2020) Indian medicinal herbs and formula-
tions for Alzheimer’s disease, from traditional knowledge to scientific assessment. Brain Sci.
https://doi.org/10.3390/brainsci10120964
41. Puttarak P et al (2017) Effects of Centella asiatica (L.) Urb. On cognitive function and mood
related outcomes: a systematic review and meta-analysis. Sci Rep. https://doi.org/10.1038/
s41598-017-09823-9
42. Sanka N, Santhipriya N, Nadendla RR (2018) An updated review on anti-Alzheimer’s herbal
drugs. Journal of Drug Delivery and Therapeutics. https://doi.org/10.22270/jddt.v8i6.2049
43. Koynova R, Tenchov B (2017) Natural product formulations for the prevention and treatment
of Alzheimer’s disease: a patent review. Recent Pat Drug Deliv Formul. https://doi.org/10.217
4/1872211312666171207152326
44. Botchway BOA (2017) Alzheimer’s disease – the past, the present and the future. Sci J Clin
Med. https://doi.org/10.11648/j.sjcm.20170601.11
45. Bondi MW, Edmonds EC, Salmon DP (2017) Alzheimer’s disease: past, present, and future. J
Int Neuropsychol Soc. https://doi.org/10.1017/S135561771700100X
46. Edmonds EC et al (2016) Heterogeneous cortical atrophy patterns in MCI not captured by con-
ventional diagnostic criteria. Neurology. https://doi.org/10.1212/WNL.0000000000003326
Natural Products Used in the Treatment
of Autoimmune Disorder
1 Introduction
A. Saharan
MM School of Pharmacy, Maharishi Markandeshwar University, Sadopur-Ambala, Haryana,
India
M. Dhanawat
Amity Institute of Pharmacy, Amity University Haryana, Gurugram, Haryana, India
C. Parkash Dora · R. K. Sindhu
Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
I. Verma (*)
M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University),
Mullana-Ambala, Haryana, India
autoimmune diseases are chronic, they will impart a negative impact on economic,
psychological, and social causes [2]. To counterbeat all these above-mentioned fac-
tors and to alleviate the side effects of the allopathic medicines, various herbal
plants were explored in terms of their usage in highly privileged autoimmune disor-
ders, which are discussed in detail ahead. To ensure safety, therapeutic agents and
treatment regimens should be obliged to prevent patients from autoimmunity. Plant
products contain a variety of bioactive compounds with therapeutic potential due to
which they are widely used in various ailments such as inflammatory diseases, auto-
immune diseases, infectious diseases, and cancer [3, 4]. Many autoimmune disor-
ders have common symptoms, such as skin rashes and exhaustion. Swelling and
redness, difficulty concentrating, numbness and tingling in the hands and feet, hair
loss, and skin rashes all lead to the diagnosis of immune response disorders [5].
Clones Autoreactive clones may be able to escape negative selection in the thy-
mus. It is possible that not all self-antigens are represented in the thymus, or that
some antigens are not processed and presented correctly [8].
Sex In lupus, an autoimmune disorder, a constant decline in the estrogen level has
been shown, which is used to enhance autoimmunity [10].
3.1 Type 1 Diabetes
3.1.1
Emblica officinalis
3.1.2
Momordica charantia
It is also known as bitter melon or bitter gourd, and is a climber plant that is well
known for its medicinal and nutraceuticals values. The pharmacological properties
exhibited by it are antimicrobial, anti-inflammatory, and antihelminthic. It was
found to contain saponin polypoid and alkaloid momo ricin. In vivo investigations
show that its chemical ingredients have an immunostimulatory impact, with the rise
in interferon and cell-mediated immunity mechanisms [15].
3.1.3
Nigella sativa
N. sativa, commonly known as black seeds, is a dicotyledon fruit that exhibits vari-
ous therapeutic potentials such as antidiabetic, antihypertensive, anti-inflammatory,
and antihistaminic properties. It was found to escalate the antioxidant mechanism
that prevents lipid peroxidation in diabetes [16].
3.1.4
Sida cordiflia
It is also known as flannel weed that has aphrodisiac and antidiabetic properties. It
was discovered that hypoglycemic impact of flannel by activating beta cells and
decreasing oxidative stress and free radical production [17].
3.1.5
Psidium guajava
3.1.6
Gymnema sylvestre
3.1.7
Azadirachta indica
It is commonly known as neem and is well known for its number for therapeutic
benefits. It contains various phytoconstituents such as nimbin, nimbinin, and nimbi-
dinin, which impart its properties as a stimulant, antioxidant, and as a contraceptive.
1252 A. Saharan et al.
Experimental studies reveal that it is used for the production of gamma interferon
and it also stimulates cell-mediated immunostimulant activity [20].
3.1.8
Allium sativum
3.1.9
Tinospora cordifolia
3.1.10
Trigonella foenum-graecum L.
3.1.11
Panax ginseng
3.1.12
Cinnamomum cassia
3.1.13
Plantago ovata
3.1.14
Zingiber officinale
3.2 Rheumatoid Arthritis
increased risk of herpes zoster and vascular events like deep vein thrombosis (DVT)
and pulmonary thromboembolism [38].
Curcuma longa
4.1
Tripterygium wilfordii
4.2
4.2.1 Alkaloids
Alkaloids are obtained from various natural resources and they have a variety of
pharmacological activities that contribute to the management of RA [48]. Various
research studies have shown that alkaloid compounds such as sinomenine, piper-
longumine, Withania somnifera (ashwagandha), Nigella sativa, koumine
(Gelsemium elegans), berberine, oxymatrine, and trikatu prevent arthritis progres-
sion via various pathways such as T-cell proliferation, reactive oxygen species
(ROS), and downregulation of various pro-inflammatory cytokines (tumor necrosis
factor [TNF]-α, IL-6, IL-12, and NF-kB). A recent double-blind study that included
120 RA patients demonstrated that sinomenine (120 mg twice daily) taken in com-
bination with MTX for 24 weeks significantly improved inflammation in arthritic
patients [49]. Withania somnifera has also the potential to suppress NF-kB signal-
ing, pro-inflammatory cytokines, and TNF-α and to reduce RANKL levels essential
for differentiation of osteoclast. An experimental animal study has found that allyl-
isopropylacetamide (AIA) Wistar rats treated with ashwagandha (1000 mg kg/day)
for 8 days showed significantly reduced bone destruction. It might be due to the
result of inhibition of inflammatory mediators and modulating the biochemical pro-
cesses associated with rheumatoid arthritis [50].
4.2.2 Phenolic Compounds
Various studies have shown the potential of phenolic compounds like flavonoids,
resveratrol, emodin, rosmarinic acid, and mangiferin in the treatment of arthritis.
These natural compounds have the properties to modulate both the action and the
production of inflammatory mediators via inhibition of NF-kB signaling. Flavonoids,
isolated from the citrus fruits, especially bergamot juice, have the potential to treat
arthritis. A recent clinical study involving RA patients has found that pomegranate
extract (250 mg twice a day) taken for 8 weeks, resulted in a significant reduction in
disease activity measures compared to indomethacin [51].
4.2.3 Quercetin
Quercetin is a naturally occurring flavonoid that is widely found in fruits, herbs, and
vegetables. Several recent experimental studies have demonstrated that quercetin
has the potential to treat rheumatoid arthritis by reducing cyclooxygenase and
lipoxygenase expressions and modulating the activation of the NF-kB signaling
pathway [52]. A study reported that, through a double-blind placebo-controlled
1256 A. Saharan et al.
study that included 50 RA females treated with quercetin (500 mg/day) for 8 weeks,
demonstrated that quercetin significantly improved early morning stiffness, pain,
and disease activity scores in 28 joints and reduced TNF-α levels compared to pla-
cebo [53].
4.2.4 Psoriasis
Glycyrrhiza glabra
4.3
Calendula officinalis L.
4.4
It was reported that Calendula officinalis flowers contain various bioactive com-
pounds such as terpenoids, carotenoids, flavonoids, and polyunsaturated fatty acids.
The presence of these moieties showed anti-inflammatory activities in various skin
disorders [55].
Annona squamosa
4.5
It is also known as sugar apple. The chemical constituents found in it are quamo-
cenin, annotemoyin, reticulatain, and squamocin, shown to have antioxidant and
anti-inflammatory properties, which proves its effectiveness for various skin disor-
ders [56, 57].
Natural Products Used in the Treatment of Autoimmune Disorder 1257
Melaleuca alternifolia
4.6
It is also known as the tea tree. The leaves contain various oil glands and are rich in
aromatic oil containing terpinolene and cineole, which are mainly used in the treat-
ment of acne, dandruff, and cold sores. It was used to produce allergic dermatitis
with sensitization to the sesquiterpenoid fraction [58].
Momordica charantia
4.7
Silybum marianum
4.8
They are commonly known as milk thistle seeds that contain phytoconstituents such
as silibinin (silybin), silychristin, and silydianin, which trigger an outbreak of pso-
riasis; it was found that it inhibits human T-cell activation, which occurs in psoria-
sis [60].
4.8.1 Graves’ Disease
This is the most prominent disease characterized mainly by altering the function of
the thyroid gland by stimulating the overproduction of hormones. Thyroid hor-
mones are used to regulate the body’s energy consumption and help in metabolism.
Some of the herbal moieties such as isoflavonoids, rosmarinic acid, blue flag, gug-
gul, selenium, iodide, and vitamin D3 supplements have profound effects on thyroid
hormones and the hypothalamus [61]. Some of the herbal plants tabulated in Table 1
have a positive effect on thyroid functioning.
Crohn’s disease (CD) and ulcerative colitis (UC) are examples of inflammatory
bowel disease (IBD), which is a category of chronic illnesses defined by gastroin-
testinal tract inflammation [64]. Several anti-inflammatory medications including
1258 A. Saharan et al.
5.1 Multiple Sclerosis
S. Biological Active
no. source constituents Mechanism of action References
1. Ananas Bromelain Reduces mRNA expression of Verma et al.
comosus pro-inflammatory cytokines interleukin [67]
(IL)-8 and tumor necrosis factor
(TNF)-α
2. Panax Ginsenosides Inhibits inflammatory cytokines and Wang et al.
quinquefolius restores microbiome inhibited [68]
by azoxymethane/dextran sodium
sulfate (AOM/DSS)
3. Penicillium Pyrenocine A Inhibits nitrite production and the Toledo et al.
paxillin synthesis of pro-inflammatory [69]
cytokines and Prostaglandin E2 (PGE2)
4. Styrax Styraxosides A Inhibits the expression of nitric oxide Yun et al.
japonica synthase (NOS) and cyclooxygenase-2 [70]
(COX-2); mRNA expression levels of
NOS and COX-2, TNF-α, and IL-1;
and inhibits the nuclear factor-kappa B
(NF-kB) pathway’s DNA-binding
capacity
5. Euphorbia Euphol Reduces the expression of NOS2, Dutra et al.
tirucalli Vascular endothelial growth factor [71]
(VEGF), and Ki67 in colonic tissues;
inhibits the levels and expression of
IL-1, chemokine (C-X-C motif) ligand
1 (CXCL1)/KC, Monocyte
chemoattractant protein-1 (MCP-1),
Macrophage Inflammatory Protein-1
(MIP-2), TNF-α, and IL-6 in colonic
tissue
6. Cinnamomum trans- Significant reduction of the Schink et al.
verum Cinnamaldehyde Lipopolysaccharides (LPS)-dependent [72]; Yu
and p-cymene IL-8 secretion et al. [73]
alleviating clinical symptoms [74] (Table 3). Natural products used in the treatment
of multiple sclerosis has been depicted in Table 3.
6 Conclusion
Autoimmune diseases occurs because of the body’s natural defenses the immune
system - attack the body’s own healthy tissue. Establishing proper balance through
the use of above natural products, and lifestyle changes may thus offer a safe and
effective alternative to traditional treatment and bring new hope to patients who are
suffering from the autoimmune diseases. Chemical compounds isolated from
1260 A. Saharan et al.
various plants have the potential to treat diabetes, rheumatoid arthritis, psoriasis,
inflammatory bowel disease, multiple sclerosis, and ulcerative colitis. There are
thousands of texts and monographs available on herbal remedies, but a majority of
medicines are not envisaged in research due to lack of knowledge. The present lit-
erature summarized the therapeutic and medicinal benefits of various natural
compounds.
References
1. Chaplin DD (2010) Overview of the immune response. J Allergy Clin Immunol 125(2):3–23
2. Missoum H, Alami M, Bachir F et al (2019) Prevalence of autoimmune diseases and clini-
cal significance of autoantibody profile: data from National Institute of Hygiene in Rabat,
Morocco. Hum Immunol 80(7):523–532
3. Ho LJ, Lai JH (2004) Chinese herbs as immunomodulators and potential disease-modifying
antirheumatic drugs in autoimmune disorders. Curr Drug Metab 5(2):181–192
4. Srivastava S, Singh D, Patel S et al (2017) Role of enzymatic free radical scavengers in man-
agement of oxidative stress in autoimmune disorders. Int J Biol Macromol 101:502–517
5. Baizabal-Carvallo JF, Jankovic J (2015) Stiff-person syndrome: insights into a complex auto-
immune disorder. J Neurol Neurosurg Psychiatry 86(8):840–848
6. Marrack P, Kappler J, Kotzin BL (2001) Autoimmune disease: why and where it occurs. Nat
Med 7(8):899–905
7. Cooper GS, Stroehla BC (2003) The epidemiology of autoimmune diseases. Autoimmun Rev
2(3):119–125
8. Inshaw JR, Cutler AJ, Burren OS et al (2018) Approaches and advances in the genetic causes
of autoimmune disease and their implications. Nat Immunol 19(7):674–684
9. Asano M, Toda M, Sakaguchi N et al (1996) Autoimmune disease as a consequence of devel-
opmental abnormality of a T cell subpopulation. J Exp Med 184(2):387–396
10. Bruserud O, Oftedal BE, Wolff AB et al (2016) AIRE-mutations and autoimmune disease.
Curr Opin Immunol 43:8–15
Natural Products Used in the Treatment of Autoimmune Disorder 1261
36. Oldsworth EA, Donaghy B, Fox KM et al (2021) Biologic and targeted synthetic DMARD uti-
lization in the United States: Adelphi Real World disease specific programme for rheumatoid
arthritis. Rheumatol Ther 8(4):1637–1649
37. Ruderman EM (2012) Overview of safety of non-biologic and biologic DMARDs.
Rheumatology 51:37–43
38. Scott IC, Hider SL, Scott DL (2018) Thromboembolism with Janus Kinase (JAK) inhibitors
for rheumatoid arthritis: how real is the risk. Drug Saf 41(7):645–653
39. Prager N, Bickett K, French N (2002) A randomized, double-blind, placebo-controlled trial to
determine the effectiveness of botanically derived inhibitors of 5-α-reductase in the treatment
of androgenetic alopecia. Altern Complement Med 8(2):143–152
40. Zheng Z, Sun Y, Liu Z (2015) The effect of curcumin and its nanoformulation on adjuvant-
induced arthritis in rats. Drug Des Dev Ther 9:4931–4942
41. Tong KK, Yang D, Chan EY et al (1999) Downregulation of lymphocyte activity and human
synovial fibroblast growth in rheumatoid arthritis by triptolide. Drug Dev Res 47:144–153
42. Kapoor B, Gupta R, Gupta M (2012) Natural products in the treatment of rheumatoid arthritis.
Int J Green Pharm 11:356–363
43. Jeyakodi S, Nai A, Divya K et al (2021) Safety and efficacy of biosolve curcumin in active
rheumatoid arthritis: a randomized double-blind placebo-controlled clinical study. Am J
Phytomed Clin Ther 9(2):5
44. Zhu W, Li Y, Zhao J et al (2022) The mechanism of triptolide in the treatment of connective
tissue disease-related interstitial lung disease based on network pharmacology and molecular
docking. Ann Med 54(1):541–552
45. Chen SR, Dai Y, Zhao J et al (2018) A mechanistic overview of triptolide and celastrol, natural
products from Tripterygiumwilfordii Hook F. Front Pharmacol 9:104
46. Fan D, Guo Q, Shen J et al (2018) The effect of triptolide in rheumatoid arthritis: from basic
research towards clinical translation. Int J Mol Sci 19(2):376
47. Gong Y, Huang X, Wang D et al (2017) Triptolide protects bone against destruction by tar-
geting RANKL-mediated ERK/AKT signalling pathway in the collagen-induced rheumatoid
arthritis. Biomed Res 28:4111–4116
48. Santiago LÂM, Neto RNM, Ataíde ACS et al (2021) Flavonoids, alkaloids and saponinsare
these plant-derived compounds an alternative to the treatment of rheumatoid arthritis, A litera-
ture review. Clin Phytoscience 7(1):1–10
49. Huang R, Pan H d, Wu J et al (2019) Comparison of combination therapy with methotrexate
and sinomenine or leflunomide for active rheumatoid arthritis: a randomized controlled clini-
cal trial. Phytomedicine 57:403–410
50. Sultana F, Neog MK, Rasool MK (2017) Withaferin-A, a steroidal lactone encapsulated man-
nose decorated liposomes ameliorates rheumatoid arthritis by intriguing the macrophage repo-
larization in adjuvant-induced arthritic rats. Colloids Surf B: Biointerfaces 155:349–365
51. Skoczyńska M, Swierkot J (2018) The role of diet in rheumatoid arthritis. Reumatologia
56(4):259–267
52. Shen P, Lin W, Deng X et al (2021) Potential implications of quercetin in autoimmune dis-
eases. Front Immunol 23(12):199. https://doi.org/10.3389/fimmu.2021.689044
53. Javadi F, Ahmadzadeh A, Eghtesadi S et al (2017) The effect of quercetin on inflammatory
factors and clinical symptoms in women with rheumatoid arthritis: a double-blind, randomized
controlled trial. J Am Coll Nutr 36(1):9–15
54. Marino A, Paterniti I, Cordaro M et al (2015) Role of natural antioxidants and potential use of
bergamot in treating rheumatoid arthritis. Pharma Nutrition 3(2):53–59
55. Raut AS, Prabhu RH, Patravale VB (2013) Psoriasis clinical implications and treatment: a
review. Crit Rev Ther Drug Carrier Syst 30(3):183–216
56. Chandrasekar R, Sivagami B (2016) Alternative treatment for psoriasis-a review. Int J Res
5(4):2188–2197
57. Bhoir SS, Vishwapathi V, Singh KK (2019) Antipsoriatic potential of Annonasquamosa seed
oil: an in vitro and in vivo evaluation. Int J Phytomedicine 54:265–277
Natural Products Used in the Treatment of Autoimmune Disorder 1263
58. Pazyar N, Yaghoobi R (2012) Tea tree oil as a novel antipsoriasis weapon. Skin Pharmacol
Physiol 25(3):162–163
59. Sharma S, Sharma R, Goyal K et al (2021) Potential of herbal treatment of Psoriasis: a laconic
review. Asian J Res Pharm Sci 11(1):51–57
60. Daniyal M, Akram M, Zainab R et al (2019) Progress and prospects in the management of
psoriasis and developments in phyto-therapeutic modalities. Dermatol Ther 32(3):e12866
61. Ng CM, Judy PS, Shek CC et al (2012) Hyperthyroidism caused by a Grave disease that is not
Graves’ disease. Am J Med 125(3):e3–e4
62. Bhardwaj M, Kumar A, Tripathi S (2013) Commiphorawightii down regulates HMG CoA
reductase in hyperlipidemic rats. Int J Appl Pharm Sci 4:441–447
63. Chee PP, Fober KA, Slightom JL (1989) Transformation of soybean (Glycine max) by infect-
ing germinating seeds with Agrobacterium tumefaciens. Plant Physiol 91(3):1212–1218
64. Mannaa FA, Abdel-Wahhab KG, Hassan LK et al (2021) Influence of Melissa officinalis meth-
anolic extract on hyperthyroidism in a rat model. Egypt Pharm J 20(2):134
65. Yeshi K, Ruscher R, Hunter L (2020) Revisiting inflammatory bowel disease: pathology, treat-
ments, challenges and emerging therapeutics including drug leads from natural products. J
Clin Med 9(5):1273
66. Hossen I, Hua W, Ting L et al (2020) Phytochemicals and inflammatory bowel disease: a
review. Crit Rev Food Sci Nutr 60(8):1321–1345
67. Verma N, Meena NK, Majumdar I et al (2017) Role of bromelain as herbal anti-inflammatory
compound using in vitro and in vivo model of colitis. J Autoimmun 3:1–8
68. Wang CZ, Yu C, Wen XD et al (2016) American ginseng attenuates colitis-associated colon
carcinogenesis in mice: impact on gut microbiota and metabolomics. Cancer Prev Res
9(10):803–811
69. Toledo TR, Dejani NN, Monnazzi LG (2014) Potent anti-inflammatory activity of pyreno-
cine A isolated from the marine-derived fungus Penicilliumpaxilli Ma (G) K. Mediat Inflamm
2014:767061
70. Yun KJ, Min BS, Kim JY et al (2007) Styraxoside A isolated from the stem bark of Styrax
japonica inhibits lipopolysaccharide-induced expression of inducible nitric oxide synthase and
cyclooxygenase-2 in RAW 264.7 cells by suppressing nuclear factor-kappa B activation. Biol
Pharm Bull 30:139–144
71. Dutra RC, Claudino RF, Bento AF et al (2011) Preventive and therapeutic euphol treatment
attenuates experimental colitis in mice. PLoS One 6:27122
72. Schink A, Naumoska K, Kitanovski Z et al (2018) Anti-inflammatory effects of cinnamon
extract and identification of active compounds influencing the TLR2 and TLR4 signaling path-
ways. Food Funct 9:5950–5964
73. Yu S, Liu M, Hu K (2019) Natural products: potential therapeutic agents in multiple sclerosis.
Int Immunopharmacol 67:87–97
74. Selmaj I, Mycko MP, Raine CS et al (2017) The role of exosomes in CNS inflammation and
their involvement in multiple sclerosis. J Neuroimmunol 306:1–10
75. Giacoppo S, Pollastro F, Grassi G et al (2017) Target regulation of PI3K/Akt/mTOR pathway
by cannabidiol in treatment of experimental multiple sclerosis. Fitoterapia 116:77–84
76. Hashimoto M, Yamamoto S, Iwasa K et al (2017) The flavonoid Baicalein attenuates cuprizone-
induced demyelination via suppression of neuroinflammation. Brain Res Bull 135:47–52
77. Hanisch UK, Kettenmann H (2007) Microglia: active sensor and versatile effector cells in the
normal and pathologic brain. Nat Neurosci 10(11):1387–1394
78. Liu HS, Shi HL, Huang F et al (2016) Astragaloside IV inhibits microglia activation via glu-
cocorticoid receptor mediated signaling pathway. Sci Rep 6(1):1–4
Strategies to Improve Antimicrobial
Activity of Natural Products: Approaches
and Challenges
1 Introduction
The plants possess a variety of bioactive compounds, which have an array of bio-
logical activities. Many synthetic drugs have their origin from some plant-based
compounds. Most spices and herbs exert antimicrobial activity against different
microorganisms (bacteria, yeasts, and molds) and can be used as food preservatives
[1]. Besides, some plants have antimicrobial effects against human and plant patho-
gens. In the last decades, plant-based antimicrobials have received considerable
attention, because plants produce secondary metabolites in abundance, known to
possess antimicrobial activity [2]. Some of these metabolites are carbohydrates,
alkaloids, tannins, steroids, terpenoids, flavonoids, and so on [3].
But they still have numerous limitations for being used as antimicrobials [4]. The
emergence of drug-resistant and multidrug-resistant (MDR) organisms has become a
Cristina M. Pérez Zamora and Carola A. Torres contributed equally with all other contributors.
global health concern affecting the diagnosis, treatment, and prevention of infections. As
just one example, the coronavirus disease-2019 (COVID-19) pandemic is causing
numerous bacterial and fungal infections in vulnerable patients hospitalized in intensive
care units (ICUs) [5]. Predictions for the year 2050 estimate that 300 million deaths will
result from this problem, with a loss of USD 100 billion to the global economy [6]. In
parallel with increasing rates of resistance to conventional antibiotics is the slowdown in
the development of new antimicrobials by the pharmaceutical industry, which has
aggravated the situation, leaving medical professionals with no alternatives for treating
patients battling life-threatening infections. Several studies have been published in
recent years on the subject of microbial resistance and the search for new therapeutic
options [7]. One method to resolve the threat of the development of drug resistance is to
use plant-based antimicrobials or their combination with synthetic antimicrobials [8, 9].
On the other hand, essential oils (EOs) from different traditionally used plants
are becoming increasingly important in the formulation of new products, mainly as
food preservatives against fungal and aflatoxin contamination [10, 11]. Different
technological treatments facilitate the utilization of natural bioactive compounds at
lower doses with improved antimicrobial effects.
This chapter aims to describe the advances made in recent years to improve the
antimicrobial activity of natural products derived from plants, mainly against clini-
cal pathogens and food contaminants. The field of application of these findings, the
main results obtained, and the difficulties and perspectives will also be discussed.
2 Antimicrobial Combinations
Here are several possibilities of combinations to consider: (1) plant extracts and
commercial antimicrobials, (2) plant extracts with each other or with natural prod-
ucts, (3) different fractions instead of the whole extract and commercial antimicro-
bials, and (4) fractions with other natural products.
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1267
Studies with plant extracts and commercial antimicrobials have been conducted
in different parts of the world [14–49]. In all cases, synergism was reported in some
or all combinations with antibiotics. The extraction methods and solvents used vary
according to the authors consulted. This is to be expected due to the variability of
chemical compounds present in the plant, which have an affinity for solvents of dif-
ferent polarity. Variability also occurs depending on the plant organ used. In most
cases, the checkerboard technique, fractional inhibitory concentration index (FICI)
calculation, and time-kill assay are used to determine the type of antibacterial inter-
action [14–46]. Disk diffusion technique is also mentioned, but much less frequently
[47–49]. The antibiotics most commonly used were gentamicin, chloramphenicol,
and ciprofloxacin [14–38]. Nevertheless, if the group of antimicrobials is taken into
account, aminoglycosides head the ranking with gentamicin, streptomycin, and
amikacin. They are closely followed by the group of beta-lactam antibiotics: ampi-
cillin, penicillin G, and imipenem. Regarding the microorganisms used, the most
common point was that the bacterial pathogens were selected according to the
World Health Organization (WHO) priority pathogen list: ESKAPE [14–44, 46, 48,
49]. This is an acronym for the group of bacteria, encompassing both Gram-positive
and Gram-negative species, made up of Enterococcus faecium, Staphylococcus
aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aerugi-
nosa, and Enterobacter species. These bacteria are common causes of life-
threatening nosocomial infections among critically ill and immunocompromised
individuals and are characterized by potential drug resistance mechanisms. As
regards antifungal activity, combinations of the extracts with fluconazole were most
frequent, followed by nystatin and amphotericin B [24, 36–38, 41, 45]. The ATCC
strains or clinical isolates of Candida albicans were the most commonly used fun-
gal agents [24, 28, 36, 37, 41, 45].
In addition, works with combinations of plant extracts with each other or with
natural products like terpenes or phenolic compounds were also found [50–58].
Here again, the checkerboard method appears most frequently as a technique to
determine interactions [50–55]. However, the disk diffusion method is also used in
some works [56–58]. In all cases, a potentiation of the antimicrobial effect was
observed when the extracts were combined. Interestingly, not only studies against
ESKAPE group bacteria were found, but also potential applications against oral
pathogens and food contaminants. In fact, Abdulbaqi et al. [50] demonstrated that
the combination between green tea and aqueous extracts of Salvadora persica
exhibited synergistic antiplaque activity, and could be used as an active agent to
produce oral health care products. On the other hand, Chung et al. [51] showed that
the high citric acid content of citrus fruit extracts (CFEs) is likely to contribute to
the strong synergistic effect with essential oil components (EOCs), such as carva-
crol and thymol. This study provides new insight into the utility of CFEs with EOCs
to improve not only the microbiological safety of food products containing CFEs
but also their applicability as a natural antibacterial complex.
On the other hand, several authors report promising results when using different
fractions instead of the whole extract [41, 59–68]. For example, Liu et al. [59], who
worked with an ethyl acetate fraction from Pithecellobium clypearia (S20b) and
1268 C. M. Pérez Zamora et al.
In this topic there is a plethora of articles to consider [12, 49, 69–95]. Some authors
evaluate the antibacterial and antibiotic-modifying activity of EOs, alone and com-
bined. Here again, the combination with commercial antibiotics showed a synergis-
tic effect.
More recently, Freitas et al. [69] evaluated the antibacterial and antibiotic-
modifying activity of the essential oil (EO) obtained from Baccharis reticulata and
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1269
the compound α-pinene against Pseudomonas aeruginosa and other MDR bacteria.
The essential oil (EO) potentiated norfloxacin and gentamicin against all strains
evaluated. Also, α-pinene increased the activity of norfloxacin against E. coli, as
well as potentiated the activity of norfloxacin and gentamicin against S. aureus. The
antimicrobial activity of this EO is mainly attributed to the presence of terpenes,
especially of the sesquiterpenes group. In summary, when the combinations of EO
with antibiotics were analyzed, it was found that the most used antibiotics were
ciprofloxacin and gentamicin [49, 69–85]. Regarding antifungals, fluconazole and
amphotericin B were the most used [76–79, 84–88]. When analyzing the microor-
ganisms employed, the most frequently used bacteria were clinical isolates of
MRSA, and MDR Klebsiella pneumoniae, Escherichia coli, Pseudomonas aerugi-
nosa, and Acinetobacter baumannii. Clinical isolates and ATCC strains of Candida
albicans and Candida parapsilosis led the ranking of fungi evaluated.
Similar to the previous cases, several studies with combinations of EO with other
natural products were also found [96–105]. Some of these combinations have appli-
cations in food. Clemente et al. [97] evaluated the effects of cinnamon and mustard
EOs against nine food-borne bacteria. The combinatorial assays demonstrated in
most cases an additive effect. In addition, synergies for Pseudomonas putida and
E. coli OI57:H7 were shown. Therefore, introducing them in combination could be
a great way to make a natural-active packaging. More recently, Ayari et al. [100]
demonstrated that the combination of oregano and thyme EOs showed a synergistic
effect against Bacillus cereus and Paenibacillus amylolyticus bacteria. Therefore,
the authors consider that this mixture has potential application as a food preserva-
tive. On the other hand, Kim et al. [104] proved that combinations of cinnamon bark
and lemongrass EOs, cinnamon bark and thyme thymol EOs, and lemongrass and
thyme thymol EOs showed synergistic activities on Listeria monocytogenes. Once
again, the antilisterial activity was evaluated by Iseppi et al. [101]. However, this
time EOs were combined with one bacteriocin previously isolated from seafood.
This study suggests that the combination of natural compounds such as Thymus
vulgaris and Salvia officinalis EOs and bacLp17 (bacteriocin) may be a useful
approach to the control of planktonic and sessile cells of L. monocytogenes in sea-
food products.
A major limitation of working with plant extracts is the laborious isolation and
identification of bioactive metabolites. Nevertheless, research has been conducted
on antimicrobial interactions with metabolites isolated from plants or their deriva-
tives as well as with commercially available natural products [106–125]. Gentamicin
and ciprofloxacin were the most commonly used antibiotics in the combinations
[106–119], while the microdilution broth and checkerboard technique were used to
evaluate interactions. The most analyzed chemical compounds belong to the group
1270 C. M. Pérez Zamora et al.
antibiotics at their internal sites of action. Catechin, like many lipophilic flavonoids,
can also cause rupture of the plasma membrane of the microorganism [110].
Lima et al. [108] evaluated the antibacterial, antifungal, and modulatory antibi-
otic activities of gallic acid, caffeic acid, and pyrogallol. The caffeic acid was the
best, potentiating the antibacterial effect over the three groups of bacteria studied.
Regarding potentiation of the effect of fluconazole, a synergistic effect was observed
only when assayed against Candida tropicalis, with all substances.
Regarding terpenes, a synergistic action of borneol associated with ciprofloxacin
against E. coli enterotoxigenic serotypes was noted [119]. In addition, synergistic
activity of borneol with other natural products against Candida albicans was dem-
onstrated [127].
Embelin, bioactive principle of Embelia ribes, exhibited a synergistic effect with
the antifungal drug ketoconazole against four different Malassezia spp. [122]. One
study conducted by Wan et al. [126] showed that cinnamic aldehyde, isolated from
Cinnamomum cassia, and pyrazinamide clearly act synergistically against
Mycobacterium tuberculosis in the murine model. Scanning electron microscopy
showed that M. tuberculosis cells swelled, collapsed, and ruptured after treatment
with sub-inhibitory concentration of phytocompound. This implies that the antimy-
cobacterial mechanism might be associated with the disruption of the cell wall.
In summary, the antibiotics most frequently used in combinations with natural
products were aminoglycosides, followed by beta-lactams and quinolones.
Antibiotics of the aminoglycosides class act in the inhibition of protein synthesis
such that bacterial resistance mechanisms mainly involve the activation of efflux
and target modification systems [128]. Thus, the synergistic effects that are gener-
ally obtained by the association of natural products with antibiotics are related to an
increase in the influx of the drug, which alters the permeability of the cell mem-
brane, favoring the penetration of antibiotics and potentializing its effect [129].
applications, like human health [162–172], agriculture [140, 173], and antimicro-
bial activity about inert surfaces [174].
According to the material and encapsulation technique used, three main types of
nanosystems can be distinguished [175]: (1) polymeric type nanosystems (matrix
type, shell core type, nanofibers, nanotube), (2) lipid-based nanosystems (solid
nanoparticles, nanoliposomes, nanoemulsions), and (3) cyclodextrin (CD) com-
plexes. Also, a combination of these techniques can be used to obtain products or
more active materials.
The coordinative complexation of polyphenolic compounds with metal ions is
another possible alternative in the search to enhance antimicrobial activities [176,
177]. The combinations can take a wide variety of different forms, such as gels,
nanoparticles, films, and emulsions.
There are literature reviews for several nanoencapsulation techniques [175, 178–
180]. This section will not review the technological aspects of the different method-
ologies but will present a summary of the natural extracts or pure natural compounds
that have been nanoencapsulated and whose antimicrobial effect was improved.
Nanosystems with EOs are the most widely studied, and, as reflected in the discus-
sion of this section, most of the time, the antimicrobial action of the EO is enhanced
after encapsulation.
In a study carried out by Barrera-Ruiz et al. [181], cinnamon (Cinnamomum
zeylanicum), thyme (Thymus vulgaris), and Schinus molle EOs encapsulated in chi-
tosan nanoparticles had a higher antimicrobial effect than the chitosan and EOs
alone on Staphylococcus aureus ATCC 25923, Enterococcus sp., Escherichia coli
ATCC 25922, and carbapenemase-producing Pseudomonas aeruginosa and
Klebsiella pneumoniae (BLEE +). Furthermore, chitosan nanoparticles loaded with
thyme EO, incorporated into burgers, significantly reduced the count of
Enterobacteriaceae, S. aureus, and fungi, compared to free EO [161]. In contrast,
when Vafani et al. [182] developed thyme oil-loaded chitosan-gelatin nanofibers
and tested against Clostridium perfringens, significant differences were not observed
in the antimicrobial activity as compared with free thyme oil. These different results
can be affected by the type of the formulation (nanoparticle or nanofiber), bacterial
strains used, the composition of thyme oil, and experimental conditions. Also,
Eucalyptus staigeriana EO nanoencapsulated using cashew gum (CG) presented
synergistic antimicrobial activity against L. monocytogenes and Salmonella enter-
itidis [183].
On the other hand, cinnamon EO encapsulated into chitosan nanoparticles pre-
sented improved antibacterial activity compared to free EO against Pseudomonas
fluorescens, Erwinia carotovora, and E. coli [184]. In another study, when cinna-
mon EO was nanoencapsulated by using chitosan and whey protein isolate, the
antibacterial activity slightly decreased against E. coli, Pseudomonas fragi,
S. aureus, and Shewanella putrefaciens [185]. In addition, the encapsulation
enhanced the antibacterial power of Cyperus articulatus rhizome EO against
S. aureus ATCC6538 and E. coli ATCC 8739 [186], as well as EO from nettle
(Urtica dioica) against B. cereus, E. coli, L. monocytogenes, S. aureus, and
Salmonella typhi [148].
The clove EO encapsulated with chitosan showed an inhibitory activity enhanced
on E. coli, L. monocytogenes, S. aureus, and S. typhi [187]. Similarly, Lee et al.
[188] demonstrated that clove EO with chitosan and poly-c-glutamic acid had syn-
ergistic and additive effects against Streptococcus sobrinus and S. mutans, respec-
tively, before nanoencapsulation (in solution). However, after nanoencapsulation,
they displayed synergistic activity against both strains [188]. In addition, clove-oil-
loaded chitosan nanoparticles and gelatin electrospun nanofibers were evaluated
against E. coli O157:H7 biofilms on cucumbers [153]. The results show a signifi-
cant eradication effect on E. coli O157:H7 biofilms, and satisfactory antibiofilm
effect on cucumber during four days of storage at 4 °C and 12 °C.
Zabihi et al. [189] evaluated the antibacterial activity of the Allium sativum EO
and its nanoliposomal form; the sub-inhibitory concentrations (50% and 75%) had
a significantly higher inhibitory effect on Shiga toxin-2 titer (produced by E. coli
O157:H7) than its free EO.
1274 C. M. Pérez Zamora et al.
Da Cunha et al. [202] enveloped the surface of silver catfish (Rhamdia quelen),
infected with Aeromonas hydrophila, with nanocapsules containing 3.6% of
Origanum majorana EO. The antimicrobial effects of these nanocapsules were sim-
ilar to those of the pure/free form but used 50 times less EO. All fish submitted to
daily baths with EO or nanocapsules loaded with EO showed significantly higher
survival rates than the control fish, and survival rates were similar to those treated
with the positive control (Maxflor®).
E. coli and 30.4 for S. aureus, thereby providing long-term durability of the nano-
composite finished fabric. In addition, bamboo and cotton fabrics were impregnated
with bovine albumin nanocapsules containing ethanolic extracts of three plants:
Terminalia chebula fruits, Rosmarinus officinalis leaves, and Opuntia littoralis
leaves [209]. The results showed inhibition halos of 42 and 44 mm for E. coli and
S. aureus, respectively. This activity was maintained after 30 washes, decreasing to
32 and 33 mm the zone of inhibition.
of microbials during storage (seven days) at 6 ± 1 °C. On the third day of cold stor-
age, fillets had the lowest total mesophilic aerobic bacteria and yeast and mold dur-
ing the storage period. Hence, this nanofiber could extend the shelf life of fish for
three days at 0 °C [215].
that of the free EO. This effect can be related to the capacity to release the active
compound over time [222].
In another work, the antimicrobial capacity of thymol and carvacrol against
E. coli and S. aureus was improved by forming complexes in hydroxypropyl-β-
cyclodextrins compared to free EO [223]. Also, when clove oil and thymol were
encapsulated with HP-α-CD, HP-β-CD, and HP-γ-CD, they showed significant
inhibition against E. coli O157:H7 Sakai and S. aureus compared to free clove oil
and thymol [224].
When the lavender EO was encapsulated into hydroxypropyl-β-cyclodextrin, its
antimicrobial activity increased approximately threefold against E. coli, Candida
albicans, and S. aureus [225].
Cyclodextrins can be incorporated in different matrices to improve or provide
desired properties to a composite material. On this line, films formed with different
cyclodextrins impregnated with EO of clove have shown promising antimicrobial
activity, especially against Gram-positive and Gram-negative bacteria [134]. Castro
et al. [226] developed films employing nanocrystals of cellulose chemically modi-
fied with β-cyclodextrin and hydroxypropyl-β-cyclodextrin, with a selective carva-
crol loading capacity. The antibacterial activities against Bacillus subtilis were
promising.
D-limonene (L) is a natural phenolic compound with high antioxidant and anti-
microbial activity. According to most papers, limonene incorporated in β-cyclodextrin
nanosponges (CD–NS) showed higher antibacterial activity than free limonene,
when tested against Escherichia coli, Shigella flexneri, Staphylococcus aureus, and
Enterococcus sp. [227].
In addition, this beneficial effect can be used not just by health or food applica-
tions. An in vitro study carried out by Matei et al. [173] shows that inclusion com-
plexes obtained with deep eutectic solvent and polyphenols such as gallic acid,
silymarin, ferulic acid, and curcumin have promising antifungal activity against
Phytophthora cinnamomi, a phytopathogenic fungus that causes a negative eco-
nomic and environmental impact. Cai et al. [228] complexed allyl isothiocyanate (a
compound obtained from cruciferous vegetables) with two types of cyclodextrins,
methyl-β-cyclodextrin and hydroxypropyl-β-cyclodextrin, and demonstrated that
complexes can suppress the attack of brown and white rot fungi (Gloeophyllum
trabeum and Trametes versicolor) on wood, and help maintain the original appear-
ance of wood.
Curcumin encapsulated within hydroxypropyl-β-cyclodextrin load in a hydrogel
matrix demonstrated significant antimicrobial activity against S. aureus [165].
Subsequently, silver nanoparticles (AgNP) using an aqueous solution of
curcumin:hydroxypropyl-β-cyclodextrin were developed by Gupta et al. [167].
These nanoparticles presented significant antibacterial activity against
Gluconacetobacter xylinus ATCC 23770, Pseudomonas aeruginosa NCIMB 8295,
and S. aureus NCIMB 6571.
When complexes of hinokitiol (HT) with α-cyclodextrin, β-cyclodextrin, or
γ-cyclodextrin were evaluated against Bacillus subtilis, Staphylococcus aureus,
Escherichia coli, Pseudomonas aeruginosa, Candida albicans, and Aspergillus
1280 C. M. Pérez Zamora et al.
Metallogels are gels that contain interactions binding metal and can magnify the
properties of the metal complexes [232]. Anh et al. [163] developed antibacterial
metallogels including tannic acid (TA), with pH- and H2O2-responsive properties, as
dressings for infected wounds, with different degrees of activity against S. epider-
midis, MRSA, and E. coli.
Nanoparticles of CuO using aqueous extract of Catha edulis leaf showed good
antibacterial activity against S. typhimurium [133]. A similar effect was observed
when nanoparticles of CuO were developed by using several plant extracts, such as
Acalypha indica [233], Phyllanthus amarus [234], Terminalia arjuna [235], Malva
sylvestris [236], and Gloriosa superba [237]. Also, ZnO nanoparticles with Bauhinia
tomentosa leaf extract showed improved inhibitory activity against P. aeruginosa
and E. coli [238].
At the same time, Jain et al. [239] synthesized silver nanoparticles using leaf
extracts of tulsi (Ocimum sanctum) with enhanced antibacterial properties against
E. coli.
On the other hand, an aqueous extract of saffron (Crocus sativus) wastages was
combined with silver nanoparticles (AgNP). They were tested against E. coli ATCC
25922, P. aeruginosa ATCC 27583, K. pneumoniae ATCC 9997, S. aureus ATCC
29213, B. subtilis ATCC 6633, and Shigella flexneri ATCC 12022. The unloaded
nanoparticles and free extract were not active against the microorganisms tested, but
the loaded AgNP was active with a relevant MIC value (250 μg/mL) for most of the
tested microorganisms [240].
Zinc silibinin complex [Zn(sil)(H2O)2] and mixed ligand zinc complexes such
as Zn(silibinin)(phenanthroline) [Zn(sil)(phen)] and Zn(silibinin)(neocuproine)
[Zn(sil)(neo)] have been synthesized by Vimalraj et al. [169], and demonstrated
good antibacterial activity against E. coli and S. aureus strains. The complexes
showed significant growth inhibition at 5 ug/mL against both strains compared to
control. Furthermore, the microbial inhibition produced by the complexes was bet-
ter than that produced by silibinin alone.
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1281
References
1. Gottardi D, Bukvicki D, Prasad S, Tyagi AK (2016) Beneficial effects of spices in food pres-
ervation and safety. Front Microbiol 7:1394. https://doi.org/10.3389/fmicb.2016.01394
2. Aminzare M, Abbasi Z, Amiri E, Hashemi M, Raeisi M, Mousavi N, Hassanzadazar H (2017)
Colibacillosis phytotherapy: an overview on the most important world medicinal plants effec-
tive on Escherichia coli. J Pharm Sci 9(5):629
3. Paiva PM, Gomes FS, Napoleão TH, Sá RA, Correia MTS, Coelho LCBB (2010)
Antimicrobial activity of secondary metabolites and lectins from plants. In: Méndez-Vilas A
(ed) Current research, technology and education topics in applied microbiology and micro-
bial biotechnology. Formatex Research Center, Badajoz
4. Kongkham B, Prabakaran D, Puttaswamy H (2020) Opportunities and challenges in managing
antibiotic resistance in bacteria using plant secondary metabolites. Fitoterapia 147:104762.
https://doi.org/10.1016/j.fitote.2020.104762
5. Gangneux JP, Bougnoux ME, Dannaoui E, Cornet M, Zahar JR (2020) Invasive fungal dis-
eases during COVID-19: we should be prepared. J Mycol Med 30(2):100971. https://doi.
org/10.1016/j.mycmed.2020.100971
6. Dadgostar P (2019) Antimicrobial resistance: implications and costs. Infect Drug Resist
12:3903–3910. https://doi.org/10.2147/IDR.S234610
7. Fatima H, Goel N, Sinha R, Khare SK (2021) Recent strategies for inhibiting multidrug-
resistant and β-lactamase producing bacteria: a review. Colloids Surf B Biointerfaces
205:111901. https://doi.org/10.1016/j.colsurfb.2021.111901
8. Alibi S, Crespo D, Navas J (2021) Plant-derivatives small molecules with antibacterial activ-
ity. Antibiotics 10(3):231. https://doi.org/10.3390/antibiotics10030231
9. Álvarez-Martínez FJ, Barrajón-Catalán E, Herranz-López M, Micol V (2021) Antibacterial
plant compounds, extracts and essential oils: an updated review on their effects and
putative mechanisms of action. Phytomedicine 90:153626. https://doi.org/10.1016/j.
phymed.2021.153626
10. Dwivedy AK, Singh VK, Prakash B, Dubey NK (2018) Nanoencapsulated Illicium verum
Hook. f. essential oil as an effective novel plant-based preservative against aflatoxin B1
production and free radical generation. Food Chem Toxicol 111:102–113. https://doi.
org/10.1016/j.fct.2017.11.007
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1283
11. Kumar A, Singh PP, Gupta V, Prakash B (2020) Assessing the antifungal and aflatoxin B1
inhibitory efficacy of nanoencapsulated antifungal formulation based on combination of
Ocimum spp. essential oils. Int J Food Microbiol 330:108766. https://doi.org/10.1016/j.
ijfoodmicro.2020.108766
12. Lahmar A, Bedoui A, Mokdad-Bzeouich I, Dhaouifi Z, Kalboussi Z, Cheraif I, Ghedira K,
Chekir-Ghedira L (2017) Reversal of resistance in bacteria underlies synergistic effect of
essential oils with conventional antibiotics. Microb Pathog 106:50–59
13. Wagner H, Efferth T (2017) Introduction: novel hybrid combinations containing synthetic or
antibiotic drugs with plant-derived phenolic or terpenoid compounds. Phytomedicine 37:1–3.
https://doi.org/10.1016/j.phymed.2017.10.020
14. Cushri S, Siriyong T, Na-Phatthalung P, Voravuthikunchai SP (2014) Synergistic effects
of ethnomedicinal plants of Apocynaceae family and antibiotics against clinical isolates
of Acinetobacter baumannii. Asian Pac J Trop Med 7(6):456–461. https://doi.org/10.1016/
S1995-7645(14)60074-2
15. Choi J-G, Choi J-Y, Mun S-H, Kang O-H, Bharaj P, Shin D-W, Kwon D-Y (2015)
Antimicrobial activity and synergism of Sami-Hyanglyun-Hwan with ciprofloxacin against
methicillin-resistant Staphylococcus aureus. Asian Pac J Trop Med 8(7):538–542. https://doi.
org/10.1016/j.apjtm.2015.06.010
16. De Aquino PEA, Pereira NLF, Figueredo FG, Ferreira SS, Leandro LMG, Souza JCCO et al
(2015) The association between drugs and herbal products: in vitro enhancement of the anti-
biotic activity by extracts of dry floral buttons of Egletes viscosa L. (macela). Eur J Integr
Med 7(3):258–262. https://doi.org/10.1016/j.eujim.2015.03.001
17. Ahumada-Santos YP, Soto-Sotomayor ME, Báez-Flores ME, Díaz-Camacho SP, López-
Angulo G, Eslava-Campos CA, Delgado-Vargas F (2016) Antibacterial synergism of
Echeveria subrigida (BL Rob & Seaton) and commercial antibiotics against multidrug resis-
tant Escherichia coli and Staphylococcus aureus. Eur J Integr Med 8(5):638–644. https://doi.
org/10.1016/j.eujim.2016.08.160
18. Cristo JS, Matias EF, Figueredo FG, Santos JF, Pereira NL, Junior JG et al (2016) HPLC pro-
file and antibiotic-modifying activity of Azadirachta indica A. Juss, (Meliaceae). Ind Crop
Prod 94:903–908. https://doi.org/10.1016/j.indcrop.2016.10.001
19. Mahomoodally MF, Dilmohamed S (2016) Antibacterial and antibiotic potentiating activ-
ity of Vangueria madagascariensis leaves and ripe fruit pericarp against human pathogenic
clinical bacterial isolates. J Tradit Complement Med 6(4):399–403. https://doi.org/10.1016/j.
jtcme.2015.09.002
20. Morais-Braga MFB, Sales DL, dos Santos SF, Chaves TP, Bitu VDCN, Avilez WMT et al
(2016) Psidium guajava L. and Psidium brownianum Mart ex DC. potentiate the effect of anti-
biotics against Gram-positive and Gram-negative bacteria. Eur J Integr Med 8(5):683–687.
https://doi.org/10.1016/j.eujim.2016.07.001
21. Taukoorah U, Lall N, Mahomoodally F (2016) Piper betle L. (betel quid) shows bacterio-
static, additive, and synergistic antimicrobial action when combined with conventional anti-
biotics. S Afr J Bot 105:133–140. https://doi.org/10.1016/j.sajb.2016.01.006
22. Akinyele TA, Igbinosa EO, Akinpelu DA, Okoh AI (2017) In vitro assessment of the syn-
ergism between extracts of Cocos nucifera husk and some standard antibiotics. Asian Pac J
Trop Biomed 7(4):306–313. https://doi.org/10.1016/j.apjtb.2016.12.022
23. Alves Ribeiro D, Soares Damasceno S, Augusti Boligon A, Alencar de Menezes IR, de
Almeida Souza MM, Martins da Costa JG (2017) Chemical profile and antimicrobial activ-
ity of Secondatia floribunda A. DC (Apocynaceae). Asian Pac J Trop Biomed 7(8):739–749.
https://doi.org/10.1016/j.apjtb.2017.07.009
24. Costa AR, de Lima SJ, Lima KRR, Rocha MI, Barros LM, da Costa JGM et al (2017)
Rhaphiodon echinus (Nees & Mart.) Schauer: chemical, toxicological activity and increased
antibiotic activity of antifungal drug activity and antibacterial. Microb Pathog 107:280–286.
https://doi.org/10.1016/j.micpath.2017.04.001
1284 C. M. Pérez Zamora et al.
25. de Sousa SZ, Macêdo NS, de Freitas TS, da Silva ARP, dos Santos JFS, Morais-Braga
MFB et al (2017) Antibacterial enhancement of antibiotic activity by Enterolobium contor-
tisiliquum (Vell.) Morong. Asian Pac J Trop Biomed 7(10):945–949. https://doi.org/10.1016/j.
apjtb.2017.09.006
26. Torres CA, Nuñez MB, Isla MI, Castro MP, González AM, Zampini IC (2017) Antibacterial
synergism of extracts from climbers belonging to Bignoniaceae family and commercial
antibiotics against multi-resistant bacteria. J Herb Med 8:24–30. https://doi.org/10.1016/j.
hermed.2017.02.002
27. Alayande KA, Pohl CH, Ashafa AOT (2018) Significance of combination therapy between
Euclea crispa (Thunb.) (leaf and stem bark) extracts and standard antibiotics against drug
resistant bacteria. S Afr J Bot 118:203–208. https://doi.org/10.1016/j.sajb.2018.07.025
28. Seebaluck-Sandoram R, Lall N, Fibrich B, Van Staden AB, Mahomoodally F (2018) Antibiotic-
potentiating activity, phytochemical profile, and cytotoxicity of Acalypha integrifolia Willd.
(Euphorbiaceae). J Herb Med 11:53–59. https://doi.org/10.1016/j.hermed.2017.03.005
29. Siebra ALA, Oliveira LR, Martins AO, Siebra DC, Albuquerque RS, Lemos ICS et al
(2018) Potentiation of antibiotic activity by Passiflora cincinnata Mast. front of strains
Staphylococcus aureus and Escherichia coli. Saudi J Biol Sci 25(1):37–43. https://doi.
org/10.1016/j.sjbs.2016.01.019
30. Vambe M, Aremu AO, Chukwujekwu JC, Finnie JF, Van Staden J (2018) Antibacterial
screening, synergy studies and phenolic content of seven South African medicinal plants
against drug-sensitive and-resistant microbial strains. S Afr J Bot 114:250–259. https://doi.
org/10.1016/j.sajb.2017.11.011
31. Wang N, Chen H, Xiong L, Liu X, Li X, An Q, Wang W (2018) Phytochemical profile of
ethanolic extracts of Chimonanthus salicifolius S. Y Hu leaves and its antimicrobial and
antibiotic-mediating activity. Ind Crops Prod 125:328–334. https://doi.org/10.1016/j.
indcrop.2018.09.021
32. Aelenei P, Luca SV, Horhogea CE, Rimbu CM, Dimitriu G, Macovei I et al (2019) Morus alba
leaf extract: metabolite profiling and interactions with antibiotics against Staphylococcus spp.
including MRSA. Phytochem Lett 31:217–224. https://doi.org/10.1016/j.phytol.2019.04.006
33. Andrade JC, Silva ARP, Santos ATL, Freitas MA, Carneiro JNP, Gonçalo MIP et al (2019)
UPLC-MS-ESI-QTOF characterization and evaluation of the antibacterial and modulatory
antibiotic activity of Ziziphus joazeiro Mart. aqueous extracts. S Afr J Bot 123:105–112.
https://doi.org/10.1016/j.sajb.2019.02.001
34. Blonck B, Cock IE (2019) Interactive antimicrobial and toxicity profiles of Pittosporum
angustifolium Lodd. extracts with conventional antimicrobials. J Integr Med 17:261–272.
https://doi.org/10.1016/j.joim.2019.03.006
35. Ilanko P, McDonnell PA, van Vuuren S, Cock IE (2019) Interactive antibacterial profile of
Moringa oleifera Lam. extracts and conventional antibiotics against bacterial triggers of some
autoimmune inflammatory diseases. S Afr J Bot 124:420–435. https://doi.org/10.1016/j.
sajb.2019.04.008
36. Omokhua AG, Ondua M, van Staden J, McGaw LJ (2019) Synergistic activity of extracts
of three South African alien invasive weeds combined with conventional antibiotics against
selected opportunistic pathogens. S Afr J Bot 124:251–257. https://doi.org/10.1016/j.
sajb.2019.05.023
37. Rolta R, Sharma A, Sourirajan A, Mallikarjunan PK, Dev K (2021) Combination between
antibacterial and antifungal antibiotics with phytocompounds of Artemisia annua L: a
strategy to control drug resistance pathogens. J Ethnopharmacol 266:113420. https://doi.
org/10.1016/j.jep.2020.113420
38. Rodrigues FC, dos Santos ATL, da Cruz RP, Almeida-Bezerra JW, Coutinho HDM, Ribeiro
PRV et al (2022) Antimicrobial activity, modulatory effect and phytochemical analysis of
Sida galheirensis Ulbr. (Malvaceae). S Afr J Bot 147:286–293. https://doi.org/10.1016/j.
sajb.2022.01.021
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1285
53. Maheshwari M, Althubiani AS, Abulreesh HH, Qais FA, Khan MS, Ahmad I (2019) Bioactive
extracts of Carum copticum L. enhances efficacy of ciprofloxacin against MDR enteric bacte-
ria. Saudi J Biol Sci 26(7):1848–1855. https://doi.org/10.1016/j.sjbs.2017.12.008
54. Moreno MA, Zampini IC, Isla MI (2020) Antifungal, anti-inflammatory and antioxi-
dant activity of bi-herbal mixtures with medicinal plants from Argentinean highlands. J
Ethnopharmacol 253:112642. https://doi.org/10.1016/j.jep.2020.112642
55. Quirino A, Morelli P, Capua G, Arena G, Matera G, Liberto MC, Focà A (2020) Synergistic
and antagonistic effects of Citrus bergamia distilled extract and its major components on drug
resistant clinical isolates. Nat Prod Res 34(11):1626–1629. https://doi.org/10.1080/1478641
9.2018.1522631
56. Islam R, Rahman MS, Hossain R, Nahar N, Hossin B, Ahad A, Rahman SM (2015)
Antibacterial activity of combined medicinal plants extract against multiple drug resistant
strains. Asian Pac J Trop Med 5:S151–S154. https://doi.org/10.1016/S2222-1808(15)60878-7
57. Chakraborty S, Afaq N, Singh N, Majumdar S (2018) Antimicrobial activity of
Cannabis sativa, Thuja orientalis and Psidium guajava leaf extracts against methicillin-
resistant Staphylococcus aureus. J Integr Med 16(5):350–357. https://doi.org/10.1016/j.
joim.2018.07.005
58. Archana H, Bose VG (2022) Evaluation of phytoconstituents from selected medicinal plants
and its synergistic antimicrobial activity. Chemosphere 287:132276. https://doi.org/10.1016/j.
chemosphere.2021.132276
59. Liu C, Huang H, Zhou Q, Liu B, Wang Y, Li P et al (2019) Antibacterial and antibiotic
synergistic activities of the extract from Pithecellobium clypearia against clinically impor-
tant multidrug-resistant gram-negative bacteria. Eur J Integr Med 32:100999. https://doi.
org/10.1016/j.eujim.2019.100999
60. Barbosa MF, Miranda PH, Souza CA, Ramos CS, Melo AL, Rocha JE et al (2021) Effect
of hybrid combinations of Erythroxylum revolutum Mart. leaf ethanolic extract or alkaloid-
enriched fraction with antibiotic drugs against multidrug-resistant bacteria strains.
Phytomedicine Plus 1(4):100105. https://doi.org/10.1016/j.phyplu.2021.100105
61. Lavor AKL, Matias EF, Alves EF, Santos BS, Figueredo FG, Lima LF et al (2014) Association
between drugs and herbal products: in vitro enhancement of the antibiotic activity by frac-
tions from leaves of Croton campestris A. (Euphorbiaceae). Eur J Integr Med 6(3):301–306.
https://doi.org/10.1016/j.eujim.2014.03.002
62. Matias EFF, Alves EF, do Nascimento Silva MK, de Alencar Carvalho VR, Medeiros CR, dos
Santos FAV et al (2015) Phytochemical characterization by HPLC and evaluation of antibac-
terial and aminoglycoside resistance-modifying activity of chloroform fractions of Cordia
verbenacea DC leaf extracts. Eur J Integr Med 7(3):251–257. https://doi.org/10.1016/j.
eujim.2015.03.007
63. da Silva JB, de Bessa ME, Mayorga OAS, Andrade VT, da Costa YFG, de Freitas Mendes R
et al (2018) A promising antibiotic, synergistic and antibiofilm effects of Vernonia conden-
sata Baker (Asteraceae) on Staphylococcus aureus. Microb Pathog 123:385–392. https://doi.
org/10.1016/j.micpath.2018.07.031
64. Demgne OMF, Damen F, Fankam AG, Guefack MGF, Wamba BE, Nayim P et al (2021)
Botanicals and phytochemicals from the bark of Hypericum roeperianum (Hypericaceae) had
strong antibacterial activity and showed synergistic effects with antibiotics against multidrug-
resistant bacteria expressing active efflux pumps. J Ethnopharmacol 277:114257. https://doi.
org/10.1016/j.jep.2021.114257
65. Rolta R, Kumar V, Sourirajan A, Upadhyay NK, Dev K (2020) Bioassay guided fractionation
of rhizome extract of Rheum emodi wall as bio-availability enhancer of antibiotics against
bacterial and fungal pathogens. J Ethnopharmacol 257:112867. https://doi.org/10.1016/j.
jep.2020.112867
66. Dantas-Medeiros R, Zanatta AC, de Souza LBFC, Fernandes JM, Amorim-Carmo B, Torres-
Rêgo M et al (2021) Antifungal and antibiofilm activities of b-type oligomeric procyani-
dins from commiphora leptophloeos used alone or in combination with fluconazole against
Candida spp. Front Microbiol 27. https://doi.org/10.3389/fmicb.2021.613155
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1287
81. Dumlupinar B, Karatoprak GŞ, Celik DD, Gürer ÜS, Demirci B, Gürbüz B et al (2020)
Synergic potential of Pelargonium endlicherianum Fenzl. Essential oil and antibiotic combi-
nations against Klebsiella pneumoniae. S Afr J Bot 135:117–126. https://doi.org/10.1016/j.
sajb.2020.08.022
82. Jugreet BS, Mahomoodally MF (2020) Essential oils from 9 exotic and endemic medicinal
plants from Mauritius shows in vitro antibacterial and antibiotic potentiating activities. S Afr
J Bot 132:355–362. https://doi.org/10.1016/j.sajb.2020.05.001
83. Jugreet BS, Mahomoodally MF (2021) Reprint of: essential oils from 9 exotic and endemic
medicinal plants from Mauritius show in vitro antibacterial and antibiotic potentiating activi-
ties. S Afr J Bot 140:478–485. https://doi.org/10.1016/j.sajb.2021.07.005
84. Braga AL, da Cruz RP, Carneiro JNP, dos Santos ATL, Sales DL, Bezerra CF et al (2021)
Piper regnellii (Miq.) C. DC.: chemical composition, antimicrobial effects, and modulation of
antimicrobial resistance. S Afr J Bot 142:495–501. https://doi.org/10.1016/j.sajb.2021.07.017
85. Brahim MAS, Fadli M, Hassani L, Boulay B, Markouk M, Bekkouche K et al (2015)
Chenopodium ambrosioides var. ambrosioides used in Moroccan traditional medicine can
enhance the antimicrobial activity of conventional antibiotics. Ind Crop Prod 71:37–43.
https://doi.org/10.1016/j.indcrop.2015.03.067
86. Göger G, Demirci B, Ilgın S, Demirci F (2018) Antimicrobial and toxicity profiles evaluation
of the Chamomile (Matricaria recutita L.) essential oil combination with standard antimi-
crobial agents. Ind Crop Prod 120:279–285. https://doi.org/10.1016/j.indcrop.2018.04.024
87. Khoury M, El Beyrouthy M, Ouaini N, Eparvier V, Stien D (2019) Hirtellina lobelii
DC. essential oil, its constituents, its combination with antimicrobial drugs and its mode of
action. Fitoterapia 133:130–136. https://doi.org/10.1016/j.fitote.2019.01.001
88. Jafri H, Ahmad I (2020) Thymus vulgaris essential oil and thymol inhibit biofilms and
interact synergistically with antifungal drugs against drug resistant strains of Candida
albicans and Candida tropicalis. J Mycol Med 30(1):100911. https://doi.org/10.1016/j.
mycmed.2019.100911
89. Barreto HM, de Lima IS, Coelho KMRN, Osório LR, de Almeida MR, dos Santos BHC et al
(2014) Effect of Lippia origanoides HBK essential oil in the resistance to aminoglycosides
in methicillin resistant Staphylococcus aureus. Eur J Integr Med 6(5):560–564. https://doi.
org/10.1016/j.eujim.2014.03.011
90. Kasrati A, Jamali CA, Fadli M, Bekkouche K, Hassani L, Wohlmuth H et al (2014)
Antioxidative activity and synergistic effect of Thymus saturejoides Coss. essential oils
with cefixime against selected food-borne bacteria. Ind Crop Prod 61:338–344. https://doi.
org/10.1016/j.indcrop.2014.07.024
91. Aguiar JJ, Sousa CP, Araruna MK, Silva MK, Portelo AC, Lopes JC (2015) Antibacterial and
modifying-antibiotic activities of the essential oils of Ocimum gratissimum L. and Plectranthus
amboinicus L. Eur J Integr Med 7(2):151–156. https://doi.org/10.1016/j.eujim.2014.10.005
92. Sienkiewicz M, Łysakowska M, Kowalczyk E, Szymańska G, Kochan E, Krukowska J et al
(2017) The ability of selected plant essential oils to enhance the action of recommended anti-
biotics against pathogenic wound bacteria. Burns 43(2):310–317. https://doi.org/10.1016/j.
burns.2016.08.032
93. Salem N, Kefi S, Tabben O, Ayed A, Jallouli S, Feres N et al (2018) Variation in chemical
composition of Eucalyptus globulus essential oil under phenological stages and evidence syn-
ergism with antimicrobial standards. Ind Crop Prod 124:115–125. https://doi.org/10.1016/j.
indcrop.2018.07.051
94. Sharma K, Guleria S, Razdan VK, Babu V (2020) Synergistic antioxidant and antimicrobial
activities of essential oils of some selected medicinal plants in combination and with synthetic
compounds. Ind Crop Prod 154:112569. https://doi.org/10.1016/j.indcrop.2020.112569
95. Rocha RR, Matos MNC, Guerrero JAP, Cavalcante RMB, Melo RS, Azevedo ÁMA et al
(2021) Comparative study of the chemical composition, antibacterial activity and synergic
effects of the essential oils of Croton tetradenius baill. and C. pulegiodorus baill. Against
Staphylococcus aureus isolates. Microb Pathog 156:104934. https://doi.org/10.1016/j.
micpath.2021.104934
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1289
96. Goldbeck JC, do Nascimento JE, Jacob RG, Fiorentini ÂM, da Silva WP (2014) Bioactivity
of essential oils from Eucalyptus globulus and Eucalyptus urograndis against planktonic cells
and biofilms of Streptococcus mutans. Ind Crop Prod 60:304–309. https://doi.org/10.1016/j.
indcrop.2014.05.030
97. Clemente I, Aznar M, Silva F, Nerín C (2016) Antimicrobial properties and mode of action of
mustard and cinnamon essential oils and their combination against foodborne bacteria. Innov
Food Sci Emerg Technol 36:26–33. https://doi.org/10.1016/j.ifset.2016.05.013
98. Veras HN, Rodrigues FF, Botelho MA, Menezes IR, Coutinho HD, Costa JG (2017)
Enhancement of aminoglycosides and β-lactams antibiotic activity by essential oil of Lippia
sidoides Cham. and the thymol. Arab J Chem 10:S2790–S2795. https://doi.org/10.1016/j.
arabjc.2013.10.030
99. Vázquez-Sánchez D, Galvão JA, Mazine MR, Gloria EM, Oetterer M (2018) Control of
Staphylococcus aureus biofilms by the application of single and combined treatments
based in plant essential oils. Int J Food Microbiol 286:128–138. https://doi.org/10.1016/j.
ijfoodmicro.2018.08.007
100. Ayari S, Shankar S, Follett P, Hossain F, Lacroix M (2020) Potential synergistic antimi-
crobial efficiency of binary combinations of essential oils against Bacillus cereus and
Paenibacillus amylolyticus - Part A. Microb Pathog 141:104008. https://doi.org/10.1016/j.
micpath.2020.104008
101. Iseppi R, Camellini S, Sabia C, Messi P (2020) Combined antimicrobial use of essential oils
and bacteriocin bacLP17 as seafood biopreservative to control Listeria monocytogenes both
in planktonic and in sessile forms. Res Microbiol 171(8):351–356. https://doi.org/10.1016/j.
resmic.2020.07.002
102. Ngo-Mback MNL, Babii C, Dongmo PJ, Toghueo MK, Stefan M, Boyom FF (2020)
Anticandidal and synergistic effect of essential oil fractions from three aromatic plants used
in Cameroon. J Mycol Med 30(2):100940. https://doi.org/10.1016/j.mycmed.2020.100940
103. Chraibi M, Fadil M, Farah A, Lebrazi S, Fikri-Benbrahim K (2021) Antimicrobial com-
bined action of Mentha pulegium, Ormenis mixta and Mentha piperita essential oils against
S. aureus, E. coli and C. tropicalis: application of mixture design methodology. LWT
145:111352. https://doi.org/10.1016/j.lwt.2021.111352
104. Kim J, Kim H, Beuchat LR, Ryu JH (2021) Synergistic antimicrobial activities of plant essen-
tial oils against Listeria monocytogenes in organic tomato juice. Food Control 125:108000.
https://doi.org/10.1016/j.foodcont.2021.108000
105. Soulaimani B, El Hidar N, El Fakir SB, Mezrioui N, Hassani L, Abbad A (2021) Combined
antibacterial activity of essential oils extracted from Lavandula maroccana (Murb.), Thymus
pallidus Batt. and Rosmarinus officinalis L. against antibiotic-resistant Gram-negative bacte-
ria. Eur J Integr Med 43:101312. https://doi.org/10.1016/j.eujim.2021.101312
106. Subramaniam S, Keerthiraja M, Sivasubramanian A (2014) Synergistic antibacterial action of
β-sitosterol-D-glucopyranoside isolated from Desmostachya bipinnata leaves with antibiot-
ics against common human pathogens. Rev Bras Farmacogn 24(1):44–50. https://doi.org/1
0.1590/0102-695X20142413348
107. Albano M, Alves FCB, Andrade BFMT, Barbosa LN, Pereira AFM, de Souza MDLR et al
(2016) Antibacterial and anti-staphylococcal enterotoxin activities of phenolic compounds.
Innov Food Sci Emerg Technol 38:83–90. https://doi.org/10.1016/j.ifset.2016.09.003
108. Lima VN, Oliveira-Tintino CD, Santos ES, Morais LP, Tintino SR, Freitas TS et al (2016)
Antimicrobial and enhancement of the antibiotic activity by phenolic compounds: gal-
lic acid, caffeic acid and pyrogallol. Microb Pathog 99:56–61. https://doi.org/10.1016/j.
micpath.2016.08.004
109. Zuo GY, Wang CJ, Han J, Li YQ, Wang GC (2016) Synergism of coumarins from
the Chinese drug Zanthoxylum nitidum with antibacterial agents against methicillin-
resistant Staphylococcus aureus (MRSA). Phytomedicine 23(14):1814–1820. https://doi.
org/10.1016/j.phymed.2016.11.001
1290 C. M. Pérez Zamora et al.
110. Gomes FMS, da Cunha XJ, Dos Santos JFS, de Matos YMLS, Tintino SR, de Freitas
TS, Coutinho HDM (2018) Evaluation of antibacterial and modifying action of catechin
antibiotics in resistant strains. Microb Pathog 115:175–178. https://doi.org/10.1016/j.
micpath.2017.12.058
111. Bustos PS, Deza-Ponzio R, Páez PL, Cabrera JL, Virgolini MB, Ortega MG (2018) Flavonoids
as protective agents against oxidative stress induced by gentamicin in systemic circula-
tion. Potent protective activity and microbial synergism of luteolin. Food Chem Toxicol
118:294–302. https://doi.org/10.1016/j.fct.2018.05.030
112. Torres CA, Sturla MA, Romero AM, Judis MA (2019) Bioguided isolation of antimicrobial
polyphenols from Cuspidaria convoluta leaves and their synergistic effect with antibiotics.
Asian Pac J Trop Biomed. https://doi.org/10.4103/2221-1691.269525
113. Zuo GY, Yang CX, Han J, Li YQ, Wang GC (2018) Synergism of prenylflavonoids from
Morus alba root bark against clinical MRSA isolates. Phytomedicine 39:93–99. https://doi.
org/10.1016/j.phymed.2017.12.023
114. Bhattacharya S, Sen D, Bhattacharjee C (2019) In vitro antibacterial effect analysis of sta-
bilized PEGylated allicin-containing extract from Allium sativum in conjugation with other
antibiotics. Process Biochem 87:221–231. https://doi.org/10.1016/j.procbio.2019.09.025
115. Parvez MAK, Saha K, Rahman J, Munmun RA, Rahman MA, Dey SK et al (2019)
Antibacterial activities of green tea crude extracts and synergistic effects of epigallocat-
echingallate (EGCG) with gentamicin against MDR pathogens. Heliyon 5(7). https://doi.
org/10.1016/j.heliyon.2019.e02126
116. Ušjak D, Ivković B, Božić DD, Bošković L, Milenković M (2019) Antimicrobial activity of
novel chalcones and modulation of virulence factors in hospital strains of Acinetobacter bau-
mannii and Pseudomonas aeruginosa. Microb Pathog 131:186–196. https://doi.org/10.1016/j.
micpath.2019.04.015
117. Cruz BG, Dos Santos HS, Bandeira PN, Rodrigues THS, Matos MGC, Nascimento MF et al
(2020) Evaluation of antibacterial and enhancement of antibiotic action by the flavonoid
kaempferol 7-O-β-D-(6″-O-cumaroyl)-glucopyranoside isolated from Croton piauhiensis
müll. Microb Pathog 143. https://doi.org/10.1016/j.micpath.2020.104144
118. Siqueira MMR, Freire PDTC, Cruz BG, de Freitas TS, Bandeira PN, Dos Santos HS et al
(2021) Aminophenyl chalcones potentiating antibiotic activity and inhibiting bacterial efflux
pump. Eur J Pharm Sci 158. https://doi.org/10.1016/j.ejps.2020.105695
119. Leite-Sampaio NF, Gondim CNF, de Souza CES, Coutinho HD (2022) Antibiotic potentiat-
ing action of α-PINENE and borneol against EPEC and ETEC sorotypes. Microb Pathog 162.
https://doi.org/10.1016/j.micpath.2021.105371
120. Cabral V, Luo X, Junqueira E, Costa SS, Mulhovo S, Duarte A et al (2015) Enhancing activity
of antibiotics against Staphylococcus aureus: zanthoxylum capense constituents and deriva-
tives. Phytomedicine 22(4):469–476. https://doi.org/10.1016/j.phymed.2015.02.003
121. Lee S, Al Razqan GS, Kwon DH (2017) Antibacterial activity of epigallocatechin-3-gallate
(EGCG) and its synergism with β-lactam antibiotics sensitizing carbapenem-associated
multidrug resistant clinical isolates of Acinetobacter baumannii. Phytomedicine 24:49–55.
https://doi.org/10.1016/j.phymed.2016.11.007
122. Sivasankar C, Gayathri S, Bhaskar JP, Krishnan V, Pandian SK (2017) Evaluation of selected
Indian medicinal plants for antagonistic potential against Malassezia spp. and the synergistic
effect of embelin in combination with ketoconazole. Microb Pathog 110:66–72. https://doi.
org/10.1016/j.micpath.2017.06.026
123. Ferreira GRS, de Santana BJ, Procopio TF, de Lima Santos ND, de Lima BJRC, Coelho
LCBB et al (2018) Antimicrobial potential of Alpinia purpurata lectin (ApuL): growth
inhibitory action, synergistic effects in combination with antibiotics, and antibiofilm activity.
Microb Pathog 124:152–162. https://doi.org/10.1016/j.micpath.2018.08.027
124. da Silva PM, da Silva BR, de Oliveira Silva JN, de Moura MC, Soares T, Feitosa APS
et al (2019) Punica granatum sarcotesta lectin (PgTeL) has antibacterial activity and syn-
ergistic effects with antibiotics against β-lactamase-producing Escherichia coli. Int J Biol
135:931–939. https://doi.org/10.1016/j.ijbiomac.2019.06.011
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1291
125. Pal A, Tripathi A (2020) Demonstration of bactericidal and synergistic activity of quercetin
with meropenem among pathogenic carbapenem resistant Escherichia coli and Klebsiella
pneumoniae. Microb Pathog 143:104120. https://doi.org/10.1016/j.micpath.2020.104120
126. Wan CJ, Zhang Y, Liu CX, Yang ZC (2022) Cinnamic aldehyde, isolated from Cinnamomum
cassia, alone and in combination with pyrazinamide against Mycobacterium tuberculosis
in vitro and in vivo. S Afr J Bot 144:200–205. https://doi.org/10.1016/j.sajb.2021.08.009
127. Liu Y, Zhao Y, Guo DL, Liu WW, Liu YX (2017) Synergistic antimicrobial activity of ber-
berine hydrochloride, baicalein and borneol against Candida albicans. Chin Herb Med
9(4):353–357. https://doi.org/10.1016/S1674-6384(17)60115-1
128. Shakil S, Khan R, Zarrilli R, Khan AU (2008) Aminoglycosides versus bacteria–a description
of the action, resistance mechanism, and nosocomial battleground. J Biomed Sci 15(1):5–14.
https://doi.org/10.1007/s11373-007-9194-y
129. Cushnie TT, Lamb AJ (2011) Recent advances in understanding the antibacterial properties of
flavonoids. Int J Antimicrob 38(2):99–107. https://doi.org/10.1016/j.ijantimicag.2011.02.014
130. Delshadi R, Bahrami A, Assadpour E, Williams L, Jafari SM (2021) Nano/microencapsulated
natural antimicrobials to control the spoilage microorganisms and pathogens in different food
products. Food Control 128:108180. https://doi.org/10.1016/j.foodcont.2021.108180
131. Gupta S, Variyar PS (2016) Nanoencapsulation of essential oils for sustained release: appli-
cation as therapeutics and antimicrobials. In: Grumezescu AM (ed) Nanotechnology in
the Agri-Food Industry, Encapsulations. Academic Press. Elsevier. Pages 641–672, ISBN
9780128043073. https://doi.org/10.1016/B978-0-12-804307-3.00015-6
132. Bilia AR, Guccione C, Isacchi B, Righeschi C, Firenzuoli F, Bergonzi MC (2014) Essential
oils loaded in nanosystems: a developing strategy for a successful therapeutic approach.
Evid-Based Complement Altern Med. https://doi.org/10.1155/2014/651593
133. Gebremedhn K, Kahsay MH, Aklilu M (2019) Green synthesis of CuO nanoparticles using
leaf extract of catha edulis and its antibacterial activity. J Pharm Pharmacol 7(7):327–342.
https://doi.org/10.17265/2328-2150/2019.06.007
134. Adel AM, Ibrahim AA, El-Shafei AM, Al-Shemy MT (2019) Inclusion complex of clove
oil with chitosan/β-cyclodextrin citrate/oxidized nanocellulose biocomposite for active food
packaging. Food Packag Shelf Life 20. https://doi.org/10.1016/j.fpsl.2019.100307
135. Alderees F, Akter S, Mereddy R, Sultanbawa Y (2021) Antimicrobial activity of nanoencap-
sulated essential oils of Tasmannia lanceolata, Backhousia citriodora and Syzygium anisa-
tum against weak-acid resistant Zygosaccharomyces bailii in clear apple juice. Beverages
7(3):67. https://doi.org/10.3390/beverages7030067
136. Lee KH, Lee JS, Kim ES, Lee HG (2019) Preparation, characterization, and food applica-
tion of rosemary extract-loaded antimicrobial nanoparticle dispersions. LWT 101:138–144.
https://doi.org/10.1016/j.lwt.2018.10.072
137. Yadav A, Kujur A, Kumar A, Singh PP, Prakash B, Dubey NK (2019) Assessing the preser-
vative efficacy of nanoencapsulated mace essential oil against food borne molds, aflatoxin
B1 contamination, and free radical generation. LWT 108:429–436. https://doi.org/10.1016/j.
lwt.2019.03.075
138. Ribeiro-Santos R, Andrade M, Sanches-Silva A (2017) Application of encapsulated essential
oils as antimicrobial agents in food packaging. Curr Opin Food Sci 14:78–84. https://doi.
org/10.1016/j.cofs.2017.01.012
139. Chaudhari AK, Dwivedy AK, Singh VK, Das S, Singh A, Dubey NK (2019) Essential oils
and their bioactive compounds as green preservatives against fungal and mycotoxin contami-
nation of food commodities with special reference to their nanoencapsulation. Environ Sci
Pollut Res 26(25):25414–25431. https://doi.org/10.1007/s11356-019-05932-2
140. Singh A, Chaudhari AK, Das S, Dubey NK (2020) Nanoencapsulated Monarda citrio-
dora Cerv. ex Lag. essential oil as potential antifungal and antiaflatoxigenic agent against
deterioration of stored functional foods. J Food Sci Technol 57(8):2863–2876. https://doi.
org/10.1007/s13197-020-04318-4
1292 C. M. Pérez Zamora et al.
141. Zanetti M, Carniel TK, Dalcanton F, dos Anjos RS, Riella HG, de Araújo PH et al (2018)
Use of encapsulated natural compounds as antimicrobial additives in food packaging: a brief
review. Trends Food Sci Technol 81:51–60. https://doi.org/10.1016/j.tifs.2018.09.003
142. Zhang H, Li X, Kang H (2019) Chitosan coatings incorporated with free or nano-encapsulated
Paulownia Tomentosa essential oil to improve shelf-life of ready-to-cook pork chops. LWT
116:108580. https://doi.org/10.1016/j.lwt.2019.108580
143. Sarvinehbaghi MB, Ahmadi M, Shiran M, Azizkhani M (2021) Antioxidant and antimicrobial
activity of red onion (Allium cepa, L.) extract nanoencapsulated in native seed gums coating
and its effect on shelf-life extension of beef fillet. J Food Meas Charact 15(5):4771–4780.
https://doi.org/10.1007/s11694-021-00985-9
144. Bahrami A, Delshadi R, Assadpour E, Jafari SM, Williams L (2020) Antimicrobial-loaded
nanocarriers for food packaging applications. Adv Colloid Interf Sci 278:102140. https://doi.
org/10.1016/j.cis.2020.102140
145. Liao W, Badri W, Dumas E, Ghnimi S, Elaïssari A, Saurel R, Gharsallaoui A (2021)
Nanoencapsulation of essential oils as natural food antimicrobial agents: an overview. Appl
Sci 11(13):5778. https://doi.org/10.3390/app11135778
146. Donsì F, Annunziata M, Sessa M, Ferrari G (2011) Nanoencapsulation of essential oils to
enhance their antimicrobial activity in foods. LWT - Food Sci Technol 44(9):1908–1914.
https://doi.org/10.1016/j.lwt.2011.03.003
147. Prakash B, Kujur A, Yadav A, Kumar A, Singh PP, Dubey NK (2018) Nanoencapsulation: an
efficient technology to boost the antimicrobial potential of plant essential oils in food system.
Food Control 89:1–11
148. Bagheri R, Ariaii P, Motamedzadegan A (2021) Characterization, antioxidant and antibacte-
rial activities of chitosan nanoparticles loaded with nettle essential oil. J Food Meas Charact
15(2):1395–1402. https://doi.org/10.1007/s11694-020-00738-0
149. Lu N, Liu Y (2020) Structural, physicochemical, and functional (antioxidant-antimicrobial)
properties of 2-O-methyl-β-cyclodextrin inclusion with hexahydro-β-acids in chitosan films.
Colloids Surf B Biointerfaces 191:111002. https://doi.org/10.1016/j.colsurfb.2020.111002
150. Cui H, Wang Y, Li C, Chen X, Lin L (2021) Antibacterial efficacy of Satureja montana
L. essential oil encapsulated in methyl-β-cyclodextrin/soy soluble polysaccharide hydro-
gel and its assessment as meat preservative. LWT 152:112427. https://doi.org/10.1016/j.
lwt.2021.112427
151. Huang H, Huang C, Yin C, Khan MR, Zhao H, Xu Y et al (2020) Preparation and character-
ization of β-cyclodextrin–oregano essential oil microcapsule and its effect on storage behav-
ior of purple yam. J Sci Food Agric 100(13):4849–4857. https://doi.org/10.1002/jsfa.10545
152. Bedoya-Serna CM, Dacanal GC, Fernandes AM, Pinho SC (2018) Antifungal activity of
nanoemulsions encapsulating oregano (Origanum vulgare) essential oil: in vitro study and
application in Minas Padrão cheese. Braz J Microbiol 49:929–935. https://doi.org/10.1016/j.
bjm.2018.05.004
153. Cui H, Bai M, Rashed MM, Lin L (2018) The antibacterial activity of clove oil/chitosan
nanoparticles embedded gelatin nanofibers against Escherichia coli O157: H7 biofilms on
cucumber. Int J Food Microbiol 266:69–78. https://doi.org/10.1016/j.ijfoodmicro.2017.11.019
154. Tometri SS, Ahmady M, Ariaii P, Soltani MS (2020) Extraction and encapsulation of Laurus
nobilis leaf extract with nano-liposome and its effect on oxidative, microbial, bacterial and
sensory properties of minced beef. J Food Meas Charact 14(6):3333–3344. https://doi.
org/10.1007/s11694-020-00578-y
155. Moraes-Lovison M, Marostegan LF, Peres MS, Menezes IF, Ghiraldi M, Rodrigues RA et al
(2017) Nanoemulsions encapsulating oregano essential oil: production, stability, antibacte-
rial activity and incorporation in chicken pâté. LWT 77:233–240. https://doi.org/10.1016/j.
lwt.2016.11.061
156. Hadian M, Rajaei A, Mohsenifar A, Tabatabaei M (2017) Encapsulation of Rosmarinus offi-
cinalis essential oils in chitosan-benzoic acid nanogel with enhanced antibacterial activity in
beef cutlet against Salmonella typhimurium during refrigerated storage. LWT 84:394–401.
https://doi.org/10.1016/j.lwt.2017.05.075
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1293
190. Jemaa MB, Falleh H, Serairi R, Neves MA, Snoussi M, Isoda H, Ksouri R (2018)
Nanoencapsulated Thymus capitatus essential oil as natural preservative. Innov Food Sci
Emerg Technol 45:92–97. https://doi.org/10.1016/j.ifset.2017.08.017
191. Moghimi R, Aliahmadi A, Rafati H, Abtahi HR, Amini S, Feizabadi MM (2018) Antibacterial
and anti-biofilm activity of nanoemulsion of Thymus daenensis oil against multi-drug
resistant Acinetobacter baumannii. J Mol Liq 265:765–770. https://doi.org/10.1016/j.
molliq.2018.07.023
192. Garzoli S, Petralito S, Ovidi E, Turchetti G, Masci VL, Tiezzi A et al (2020) Lavandula x
intermedia essential oil and hydrolate: evaluation of chemical composition and antibacte-
rial activity before and after formulation in nanoemulsion. Ind Crop Prod 145. https://doi.
org/10.1016/j.indcrop.2019.112068
193. Gholipourkanani H, Buller N, Lymbery A (2019) In vitro antibacterial activity of four nano-
encapsulated herbal essential oils against three bacterial fish pathogens. Aquac 50(3):871–875.
https://doi.org/10.1111/are.13959
194. Ruiz-Gonzalez N, Lopez-Malo A, Palou E, Ramirez-Corona N, Jimenez-Munguia MT
(2019) Antimicrobial activity and physicochemical characterization of oregano, thyme and
clove leave essential oils, nonencapsulated and nanoencapsulated, using emulsification. Appl
Food Biotechnol 6(4):237–246. https://doi.org/10.22037/afb.v6i4.25541
195. Cecchini ME, Paoloni C, Campra N, Picco N, Grosso MC, Perez MS et al (2021)
Nanoemulsion of Minthostachys verticillata essential oil. In-vitro evaluation of its antibacte-
rial activity. Heliyon 7(1). https://doi.org/10.1016/j.heliyon.2021.e05896
196. Upadhyay N, Singh VK, Dwivedy AK, Chaudhari AK, Dubey NK (2021) Assessment of
nanoencapsulated Cananga odorata essential oil in chitosan nanopolymer as a green approach
to boost the antifungal, antioxidant and in situ efficacy. Int J Biol Macromol 171:480–490.
https://doi.org/10.1016/j.ijbiomac.2021.01.024
197. Izadi M, Jorf SAM, Nikkhah M, Moradi S (2021) Antifungal activity of hydrocolloid nano
encapsulated Carum copticum essential oil and Peganum harmala extract on the patho-
genic fungi Alternaria alternata. Physiol Mol Plant Pathol 116. https://doi.org/10.1016/j.
pmpp.2021.101714
198. Nirmal NP, Mereddy R, Li L, Sultanbawa Y (2018) Formulation, characterisation and anti-
bacterial activity of lemon myrtle and anise myrtle essential oil in water nanoemulsion. Food
Chem 254:1–7. https://doi.org/10.1016/j.foodchem.2018.01.173
199. Rayhane BK, Rojo SR, Gaspar F, Leila CG (2019) Preparation of rosemary essential oil-
loaded multiple lipid nanoparticles (ROMLNs) and evaluation of their antibacterial activity.
Abstract presented in at NanoSpain Conf 2019, Barcelona, Spain, 28–31 March 2019
200. Jin L, Teng J, Hu L, Lan X, Xu Y, Sheng J et al (2019) Pepper fragrant essential oil (PFEO)
and functionalized MCM-41 nanoparticles: formation, characterization, and bactericidal
activity. J Sci Food Agric 99(11):5168–5175. https://doi.org/10.1002/jsfa.9776
201. Khatibi SA, Misaghi A, Moosavy MH, Akhondzadeh Basti A, Mohamadian S, Khanjari A
(2018) Effect of nanoliposomes containing Zataria multiflora Boiss. essential oil on gene
expression of Shiga toxin 2 in Escherichia coli O157: H7. J Appl Microbiol 124(2):389–397.
https://doi.org/10.1111/jam.13641
202. da Cunha JA, de Ávila SC, Fausto VP, de Melo LDW, Henneman B, Frizzo CP et al (2018)
The antibacterial and physiological effects of pure and nanoencapsulated Origanum majo-
rana essential oil on fish infected with Aeromonas hydrophila. Microb Pathog 124:116–121.
https://doi.org/10.1016/j.micpath.2018.08.040
203. Oliveira DA, Angonese M, Ferreira SR, Gomes CL (2017) Nanoencapsulation of pas-
sion fruit by-products extracts for enhanced antimicrobial activity. Food Bioprod Process
104:137–146. https://doi.org/10.1016/j.fbp.2017.05.009
204. Pereira MC, Hill LE, Zambiazi RC, Mertens-Talcott S, Talcott S, Gomes CL (2015)
Nanoencapsulation of hydrophobic phytochemicals using poly (dl-lactide-co- glycolide)
(PLGA) for antioxidant and antimicrobial delivery applications: Guabiroba fruit
(Campomanesia xanthocarpa O. Berg) study. LWT 63(1):100–107. https://doi.org/10.1016/j.
lwt.2015.03.062
1296 C. M. Pérez Zamora et al.
205. Masjid NAS, Martien R, Dono ND (2019) In Vitro antibacterial activity of Mindi (Melia aze-
darach Linn.) leaf extract with nanoencapsulation technology. IOP Conf Ser: Earth Environ
Sci 387(1):012069. IOP Publishing
206. Nallasamy P, Ramalingam T, Nooruddin T, Shanmuganathan R, Arivalagan P, Natarajan S
(2020) Polyherbal drug loaded starch nanoparticles as promising drug delivery system: anti-
microbial, antibiofilm and neuroprotective studies. Process Biochem 92:355–364. https://doi.
org/10.1016/j.procbio.2020.01.026
207. Srivastava J, Chandra H, Nautiyal AR, Kalra SJS (2014) Antimicrobial resistance (AMR)
and plant-derived antimicrobials (PDAms) as an alternative drug line to control infections.
Biotech 4:451–460. https://doi.org/10.1007/s13205-013-0180-y
208. Chandrasekar S, Vijayakumar S, Rajendran R (2020) Functional finishing of health care cot-
ton for enhanced efficiency of antibacterial activity by chitosan and herbal nanocomposites.
Acta Ecol Sin 40(6):473–477. https://doi.org/10.1016/j.chnaes.2020.08.004
209. Banupriya J, Maheshwari V (2019) A study on antibacterial property of herbal-biopolymer
nanoencapsulate treated fabric. J Drug Deliv Ther 9(4):433–437. https://doi.org/10.22270/
jddt.v9i4.3204
210. Pan K, Chen H, Davidson PM, Zhong Q (2014) Thymol nanoencapsulated by sodium casein-
ate: physical and antilisterial properties. J Agric Food Chem 62(7):1649–1657. https://doi.
org/10.1021/jf4055402
211. de Souza TW, Pena GR, Martin-Pastor M, de Sousa FFO (2021) Design and characterization
of ellagic acid-loaded zein nanoparticles and their effect on the antioxidant and antibacterial
activities. J Mol Liq 341. https://doi.org/10.1016/j.molliq.2021.116915
212. Wang Y, Xue Y, Bi Q, Qin D, Du Q, Jin P (2021) Enhanced antibacterial activity of eugenol-
entrapped casein nanoparticles amended with lysozyme against gram-positive pathogens.
Food Chem 360:130036. https://doi.org/10.1016/j.foodchem.2021.130036
213. Oluoch G, Matiru V, Mamati EG, Nyongesa M (2021) Nanoencapsulation of thymol and
eugenol with chitosan nanoparticles and the effect against Ralstonia solanacearum. Adv
Microbiol 11(12):723–739. https://doi.org/10.4236/aim.2021.1112052
214. Lee JS, Kim ES, Lee HG (2017) Improving the water solubility and antimicrobial activity
of silymarin by nanoencapsulation. Colloids Surf B Biointerfaces 154:171–177. https://doi.
org/10.1016/j.colsurfb.2017.03.004
215. Ceylan Z, Sengor GFU, Yilmaz MT (2018) Nanoencapsulation of liquid smoke/thymol com-
bination in chitosan nanofibers to delay microbiological spoilage of sea bass (Dicentrarchus
labrax) fillets. J Food Eng 229:43–49. https://doi.org/10.1016/j.jfoodeng.2017.11.038
216. Zarzycki PK, Głód BK (2016) Cyclodextrins-based nanocomplexes for encapsulation of
bioactive compounds in food, cosmetics, and pharmaceutical products: principles of supra-
molecular complexes formation, their influence on the antioxidative properties of target
chemicals, and recent advances in selected industrial applications. In: Grumezescu AM (ed)
Encapsulations. Academic Press, Elsevier
217. Kfoury M, Auezova L, Greige-Gerges H, Fourmentin S (2019) Encapsulation in cyclodex-
trins to widen the applications of essential oils. Environ Chem Lett 17(1):129–143. https://
doi.org/10.1007/s10311-018-0783-y
218. Suvarna V, Gujar P, Murahari M (2017) Complexation of phytochemicals with cyclodex-
trin derivatives–an insight. Biomed Pharmacother 88:1122–1144. https://doi.org/10.1016/j.
biopha.2017.01.157
219. Saffarionpour S (2019) Nanoencapsulation of hydrophobic food flavor ingredients and their
cyclodextrin inclusion complexes. Food Bioprocess Technol 12(7):1157–1173. https://doi.
org/10.1007/s11947-019-02285-z
220. Ozdemir N, Pola CC, Teixeira BN, Hill LE, Bayrak A, Gomes CL (2018) Preparation of
black pepper oleoresin inclusion complexes based on beta-cyclodextrin for antioxidant and
antimicrobial delivery applications using kneading and freeze drying methods: a comparative
study. LWT 91:439–445. https://doi.org/10.1016/j.lwt.2018.01.046
Strategies to Improve Antimicrobial Activity of Natural Products: Approaches… 1297
Abbreviations
CD Cluster of differentiation
DNA Deoxyribonucleic acid
DTH Delayed-type hypersensitivity
HBD-2 Human beta-defensin-2
IL Interleukin
INF Interferon
iNOS Inducible nitric oxide synthase
L-DOPA Levodopa and l-3,4-dihydroxyphenylalanine
LFA-1-ICAM
Lymphocyte function-associated antigen -1- Intercellular
Adhesion Molecule
LPS Lipopolysaccharide
SRBC Sheep red blood cell
Th Helper T cells
THC Delta-9-tetrahydrocannabinol
TNF Tumor necrosis factor
WBC White blood cell
1 Introduction
S. P. Thekkekkoottumughath (*)
Regional Ayurveda Research Institute Thiruvananthapuram, Kerala, Under Central Council
for Research in Ayurvedic Sciences (CCRAS), Ministry of AYUSH, New Delhi, India
[1]. Extensive research activities are being carried out all over the world for the vari-
ous therapeutic potential and medicinal values of traditional remedies by the
researchers and experts who bear the responsibility of keeping the traditional sys-
tems of medicine alive.
New epidemics are evolving like the present coronavirus disease-2019
(COVID-19). The novel coronavirus (nCoV) spillover event has emerged as a pub-
lic health emergency of international concern. This is the third instance of the emer-
gence of a novel coronavirus, after severe acute respiratory syndrome (SARS) in
2003 and Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 in
the past two decades. The repeated emergence and global scale of transmission,
significant number of deaths, infection and mortality of care providers and health
care workers, and higher risk of death in vulnerable groups have been the major
causes of concern. If we have a strong immune system, we may not get afflicted by
the disease or even if we get afflicted, the manifestation of symptoms may be
delayed or occur with mild symptoms [2].
Most of the immunomodulatory drugs prescribed in conventional medicine are
reported to have various side effects like increasing the risk of infections, reactiva-
tion of latent tuberculosis, medullar suppression, gastric irritation, pancreatitis,
hepatitis, dizziness, fever, rash, alopecia, nausea, diarrhea, fatigue, elevated trans-
aminase levels, hypertension, nephropathy, convulsions, hyperkalemia, neurotoxic-
ity, hirsutism, epilepsy, headache, paresthesia, and gingival hyperplasia [3].
Ayurveda is one of the most renowned and time-tested traditional system of med-
icine, with nil or minimal side effects and has flourished from ages till date. In
future also, this system will continue to serve the humanity with unique description
of the relationship of human body constitution and function to nature and the ele-
ments of the universe that act in coordination and affect the living beings. A lot of
plants with immunomodulatory potential have been described in various Ayurvedic
classical textbooks. Different plant-based principles have been isolated from such
plants with potential immunomodulatory activities. The aim of this review is to
bring to light such plants and researches done on them. The research papers were
selected from the most pertinent databases for the biomedical sciences.
immunity is the adaptive immune reaction that develops in weeks/months and may
last throughout whole life as a result of exposure to antigen. Active immunity may
be acquired or natural. Antigen-specific reactions by B- and T lymphocytes are
involved in adaptive immunity. The strong phagocytic action of myeloid cells and
cytotoxic T lymphocytes is enhanced by helper T-cell-1 (Th1) lymphocytes. They
produce interleukin (IL)-2, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ.
Th2 lymphocytes produce IL-4, IL-5, and IL-10 and are classified by B cell facili-
tated production of antibodies. As a result of binding with antibodies, microorgan-
isms or toxins are neutralized. Additionally, through activation of complement
proteins, antibodies opsonize various pathogens, immobilize bacteria, and trigger
destruction of microorganisms by phagocytes [6].
Innate immunity includes chemical, microbiological, and physical barriers.
Macrophages, monocytes, cytokines, acute phase proteins, neutrophils, and com-
plement system are the main mediators of immune system that deliver quick defense
for body. Host recognizes various distinct moieties expressed by pathogens known
as pathogen-associated molecular patterns (PAMPs) to detect presence of a patho-
gen. They are recognized by the germline-encoded host sensors known as pattern
recognition receptors (PRRs). After that, an array of immune responses are quickly
triggered through induction of different chemokines, type I interferons, and cyto-
kines. Pattern recognition receptor families such as retinoic-acid-inducible gene I
(RIG-I)-like receptors, DNA receptors (cytosolic sensors for DNA), nucleotide-
binding oligomerization domain-like receptors (NOD-like receptors), and toll-like
receptors have a significant part in defense mechanism of the host [7].
Macrophages and antigen-presenting cells that play significant roles in secretion
of cytokines, antigen presentation, processing, antibody-dependent cell-mediated
cytotoxicity, nitric oxide (NO) production, and phagocytosis are included in all
phases of nonspecific immunity. Naive and memory B cells and naive T cells are
activated by dendritic cells. The effectors of innate immunity including natural
killer (NK) cells are regulated, which govern specific and natural immune responses
by producing granulocyte–macrophage colony-stimulating factor (GM-CSF), tumor
necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) during diverse phases of dif-
ferentiation of dendritic cells [8].
Complement system is the tertiary important element of natural immunity and is
the main effector of humoral immunity among all the physiological systems of host
defense mechanism [9].
Immunomodulators These are biological or synthetic substances that can stimu-
late, suppress, or modulate any aspect of the immune system including both adap-
tive and innate arms of the immune system. The imbalance of immune responses
may cause a plethora of morbidities, such as AIDS, allergy, autoimmune diseases,
and immunosuppression, in spite of the high adeptness and specificity of the immune
system [10, 11]. Immunomodulators have been developed as recent researches from
epidemiological studies provide evidence of a rise in immunological diseases. They
are capable of stimulating or suppressing the immune response in immune-system-
mediated diseases. Immunostimulatory drugs have been developed for managing
1302 S. P. Thekkekkoottumughath
I. Immunosuppressants
(a) Category: Inhibitors of lymphocyte gene expression
Mechanism of action: Reduce leukocyte extravasation and proinflamma-
tory cytokine expression
Example: Glucocorticoids [15, 16]
(b) Category: Inhibitors of lymphocyte signaling
Mechanism of action: Antigen-triggered signal transduction constrain in
T cells and reduced expression of lymphokines and antiapoptotic proteins
Example: Cyclosporine [16, 17]
(c) Category: Inhibitors of lymphocyte signaling
Mechanism of action: By inhibition of calcineurin, T-cell activation
Example: Tacrolimus [18, 19]
(d) Category: Inhibitors of lymphocyte signaling
Mechanism of action: Downregulate T-cell growth factor receptor and
IL-2 and inhibit activation and proliferation of T lymphocyte
Example: Sirolimus [20, 21]
(e) Category: Cytotoxic agents
Mechanism of action: Inhibit new synthesis of purine, which results in
inhibition of proliferation of lymphocytes
Examples: Azathioprine; azathioprine sodium [20]
(f) Category: Cytotoxic agents
Mechanism of action: Inhibit new synthesis of guanine by the inhibition
of inosine monophosphate dehydrogenase
Example: Mycophenolate mofetil [22, 23]
(g) Category: Alkylating agent
Mechanism of action: By cross linking in the strands of DNA, averts
protein synthesis and cell division
Example: Cyclophosphamide [24]
(h) Category: Cytokine inhibitors
Mechanism of action: Bind and inhibit TNF-α to bind with its receptors
Examples: Etanercept, infliximab, adalimumab, anakinra, daclizumab,
and basiliximab [25–27]
Plants with Immunomodulatory Potential Described in Ayurveda 1303
4.1 Immunomodulators
Fresh juice increased humoral antibody response, antibody titer in the hem-
agglutination antibody assay, and plaque-forming cell assay [88].
47. Nigella sativa L. (Family: Ranunculaceae)
Part used: Seeds
Important chemical constituents: Nigellidine and α-hederin
The proliferative capacity of splenocytes and T-lymphocytes were enhanced
and IFN-γ secretion from splenocytes was suppressed when treated with
aqueous extract of Nigella sativa [89].
48. Phyllanthus emblica L. (Family: Phyllanthaceae)
Part used: Fruits
Important chemical constituents: Polyphenols and vitamin C
B- and T-lymphocytes were stimulated and interleukin production was
restored by alcoholic extract of fruits [90].
49. Solanum nigrum L. (Family: Solanaceae)
Part used: Seeds
Important chemical constituents: Glycoalkaloids, glycoproteins, and
polysaccharides
Isolated polysaccharides of Solanum nigrum seeds showed significant incre-
ment in the percentage of CD4+ T-lymphocyte and a decrease in the percent-
age of CD8+ T-lymphocyte of peripheral blood of tumor-bearing mice [91].
50. Valeriana wallichii DC. (Family: Valerianaceae)
Part used: Roots
Important chemical constituents: Pyridine alkaloids, some organic acids,
and terpenes
Alcoholic root extract inhibited hepatitis C virus (HCV) by binding with
HCV nonstructural B (NS5B) protein [92].
51. Vitex negundo L. (Family: Verbanaceae)
Parts used: Roots and leaves
Important chemical constituents: Negundoside, agnuside, and vitegnoside
Phagocytic cells such as macrophages and neutrophils were activated by
hydroalcoholic extract of leaves of V. negundo [93].
52. Zingiber officinale Roscoe (Family: Zingiberaceae)
Part used: Rhizome
Important chemical constituents: 6-Gingerol, 6-shogaol, and 6-paradol
Cell-mediated immune response and nonspecific proliferation of
T-lymphocyte were affected by volatile oil of Zingiber officinale [94].
1314 S. P. Thekkekkoottumughath
5 Discussion
In this era of emerging pandemics like COVID-19, promising drugs to combat them
have to be discovered on an emergency basis. Antibody reagents, natural adjuvants,
and synthetic agents are used as immunomodulators in conventional medicine.
However, there are serious limitations in prescribing these medicines due to side
effects like increased risk of infection and generalized effect all over the immune
system. Consequently, there was increased usage of herbal plants, which are consid-
ered to be less toxic with fewer side effects, to treat diseases of immune system [124].
A lot of plants with immunomodulatory potential acting on various components
of immune system have been described in various Ayurvedic classical textbooks.
Different plant-based principles have been isolated from such plants with potential
immunomodulatory activity. These herbs seem to act through immunosuppressant,
immunostimulant, or immunoadjuvant activities by affecting the effector arm of the
immune response [125].
Main mechanisms of immunomodulation activity so far understood are via
immunostimulatory effect on peritoneal macrophages; cellular immune function
enhancement and nonspecific cellular immune system effect; increased nonspecific
immunity mediators; stimulation of phagocytosis; macrophage activation; lym-
phoid cell stimulation; increase in antigen-specific immunoglobulin production,
circulating total WBC count, interleukin-2, number of natural killer cells, reducing
chemotherapy-induced leukopenia, and increasing [126].
The herbal mixture preparations of Ayurveda may be acting on immune system
due to their phytochemicals with immunomodulatory properties, probably acting
synergistically [1].
From the above review it is evident that there are a lot of medicinal plants that
exert immunomodulatory activity in various experimental models. Various in vivo
and in vitro screening methods were employed to ascertain their pharmacological
Plants with Immunomodulatory Potential Described in Ayurveda 1319
activity. Some medicinal plants were observed to stimulate the immune response
(e.g., Ocimum sanctum, Tinospora cordifolia), while some suppress the immune
system (Cannabis sativa). Various secondary metabolites like sterols, alkaloids, fla-
vonoids, glycosides, saponins, and coumarins exhibit a wide range of immunomod-
ulating activity.
6 Conclusion
References
11. Lerner A, Jeremias P, Matthias T (2016) The world incidence and prevalence of autoimmune
diseases is increasing. Int J Celiac Dis 3:151–155. https://doi.org/10.12691/ijcd-3-4-8
12. Catanzaro M, Corsini E, Rosini M et al (2018) Immunomodulators inspired by nature: a
review on curcumin and echinacea. Molecules 23(11):2778. https://doi.org/10.3390/
molecules23112778
13. Dham SK (2017) Immunomodulation. Med J Armed Forces India 51(3):149–150. https://doi.
org/10.1016/S0377-1237(17)30954-1
14. Jantan I, Ahmad W, Bukhari SNA (2015) Plant-derived immunomodulators: an insight on
their preclinical evaluation and clinical trials. Front Plant Sci 6:655. https://doi.org/10.3389/
fpls.2015.00655
15. Golan DE (2008) Principles of pharmacology. The pathophysiologic basic of drug therapy.
Lippincott Williams & Wilkins, Pennsylvania, pp 795–809
16. Jain S (2008) Handbook of pharmacology. Pars Publication, London
17. Tortora GJ, Derrickson BP (2008) Principles of anatomy and physiology. Wiley, Hoboken
18. Singhal KC (2007) Essentials of pharmacotherapeutics. CBS Publishers, New Delhi
19. Sengupta PR (2009) Medical pharmacology. CBS Publishers, New Delhi
20. Katzung GB, Trevor JA (2009) Basic and clinical pharmacology. Lange Medical Books/
McGraw Hill, New York
21. Gilman’s G (2008) Manual of pharmacology and therapeutics. Professor of Pharmacology &
Medicine University of California, San Diego LaJolla
22. Rang HP, Dale MM (2007) Rang and Dale pharmacology. Churchill Livingestone, Edinburgh
23. Richard AH, Pamela CC (2009) Lippincott’s illustrated reviews: pharmacology. Wolters
Kluwer, New Delhi
24. Mythili M, Nair S, Gunasekaran S (2004) Effect of cyclophosphamide pretreatment on hema-
tological indices of Indian Bonnet monkeys. Indian J Pharmacol 36:175
25. Keane J, Gershon S, Wise RP et al (2001) Tuberculosis associated with infliximab, a
tumor necrosis factor alpha-neutralizing agent. N Engl J Med. https://doi.org/10.1056/
NEJMoa011110
26. Baudouin V, Crusiaux A, Haddad E et al (2003) Anaphylactic shock caused by immunoglobu-
lin E sensitization after retreatment with the chimeric anti-interleukin-2 receptor monoclonal
antibody basiliximab. Transplantation. https://doi.org/10.1097/01.TP.0000073809.65502.8F
27. Bartelds GM, Krieckaert CL, Nurmohamed MT et al (2011) Development of antidrug anti-
bodies against adalimumab and association with disease activity and treatment failure during
long-term follow-up. JAMA. https://doi.org/10.1001/jama.2011.406
28. Hansel TT, Kropshofer H, Singer T et al (2010) The safety and side effects of monoclonal
antibodies. Nat Rev Drug Discov. https://doi.org/10.1038/nrd3003
29. Sgro C (1995) Side-effects of a monoclonal antibody, muromonab CD3/orthoclone OKT3:
bibliographic review. Toxicology. https://doi.org/10.1016/0300-483X(95)03123-W
30. Liu L, Li Y (2014) The unexpected side effects and safety of therapeutic monoclonal antibod-
ies. Drugs Today. https://doi.org/10.1358/dot.2014.50.1.2076506
31. Berger JR, Houff SA, Major EO (2009) Monoclonal antibodies and progressive multifocal
leukoencephalopathy. MAbs. https://doi.org/10.4161/mabs.1.6.9884
32. Auffenberg C, Rosenthal LJ, Dresner N (2013) Levamisole: a common cocaine adulterant with
life-threatening side effects. Psychosomatics. https://doi.org/10.1016/j.psym.2013.02.012
33. Strayer DR, Carter WA (2012) Recombinant and natural human interferons: analysis of the
incidence and clinical impact of neutralizing antibodies. J Interferon Cytokine Res. https://
doi.org/10.1089/jir.2011.0069
34. Campoli-Richards D, Sorkin E, Heel R (1986) Inosine Pranobex. Drugs. https://doi.
org/10.2165/00003495-198632050-00001
35. Charak (2007) Deerghanjeevitheeya Adhyaya. In: Sharma RK, Bhagwan D (eds) Charaka
Samhita, 1st edn. Chaukhamba Sanskrit Series office, Varanasi
36. Vagbhata (2005) Doshadivijnaneeya. In: Srikanthamurthy KR (ed) Ashtanga Samgraha, 2nd
edn. Chaukhamba Orientalia, Varanasi
Plants with Immunomodulatory Potential Described in Ayurveda 1321
37. Charak (2008) Tisreshaniya. In: Shastri SN (ed) Charak Samhita. Chukhambha Bharty
Academy, Varanasi
38. Vagbhata (2005) Dinacharya. In: Srikanthamurthy KR (ed) Ashtanga Samgraha, 2nd edn.
Chaukhamba Orientalia, Varanasi
39. Vagbhata (2005) Ritucharya. In: Srikanthamurthy KR (ed) Ashtanga Samgraha, 2nd edn.
Chaukhamba Orientalia, Varanasi
40. Goyal M (2018) Rasayana in perspective of present scenario. Ayu 39:63–64. https://doi.
org/10.4103/ayu.AYU_300_18
41. Nadkarni KM, Nadkarni AK (2005) Indian materia medica. Popular Prakashan, Mumbai
42. Mondal S, Varma S, Bamola VD (2011) Double-blinded randomized controlled trial for
immunomodulatory effects of Tulsi (Ocimum sanctum Linn.) leaf extract on healthy volun-
teers. J Ethnopharmacol. https://doi.org/10.1016/j.jep.2011.05.012
43. Bharani SER, Asad M, Dhamanigi SS et al (2010) Immunomodulatory activity of methanolic
extract of Morus alba linn. (mulberry) leaves. Pak J Pharm Sci 23(1):63–68
44. Cooper JC, Turcasso N (1999) Immunostimulatory effects of b-1,3 glucan and acemannan.
JANA 2:5–11
45. Im SA, Lee YR, Lee YH et al (2010) In vivo evidence of the immunomodulatory activ-
ity of orally administered Aloe vera gel. Arch Pharm Res. https://doi.org/10.1007/
s12272-010-0315-1
46. Rajanna M, Bharathi B, Shivakumar BR (2021) Immunomodulatory effects of Andrographis
paniculata extract in healthy adults – an open-label study. J Ayurveda Integr Med. https://doi.
org/10.1016/j.jaim.2021.06.004
47. Gautham M, Saha S, Bani S et al (2009) Immunomodulatory activity of Asparagus rac-
emosus on systemic Th1/Th2 immunity: implications for immunoadjuvant potential. J
Ethnopharmacol. https://doi.org/10.1016/j.jep.2008.10.028
48. Paul S, Bandyopadhyay TK, Bhattacharya A (2011) Immunomodulatory effect of leaf extract
of Murraya koenigii in diabetic mice. Immunopharmacol Immunotoxicol. https://doi.org/1
0.3109/08923973.2011.561354
49. Pradhan D, Panda PK, Tripathy G (2009) Evaluation of immunomodulatory activity of meth-
anolic extract of Couroupita guianensis Aubl flowers in rat. NPR 8(1):37–42
50. Sharma U, Bala M, Kumar M et al (2012) Immunomodulatory active compounds from
Tinospora cordifolia. J Ethnopharmacol. https://doi.org/10.1016/j.jep.2012.03.027
51. Deshpande JR, Choudhary AA, Mirsha MR et al (2008) Benefical effects of Lagenaria sicer-
aria Mol. Fruit epicarp in animal models. Indian J Exp Biol 46:234–242
52. Halder S, Bharal N, Mediratta PK et al (2009) Antiinflammatory, Immunomodulatory and
anti-nociceptic activity of Terminalia arjuna Roxb. Bark powder in mice and rats. Indian J
Exp Biol 47:577–583
53. Ghaisas MM, Saikh SA, Deshpande AD (2009) Evaluation of immunomodulatory
activity of ethanolic extract of stem bark of Bauhinia variegata L. IJGP. https://doi.
org/10.4103/0973-8258.49379
54. Tekade SH, Mode SG, Waghmare SP (2008) Effect of Asparagus racemosus, Sida cordifolia
and levamisole on immunological parameters in experimentally induced immunosuppressed
broilers. Vet World 1(2):49–50
55. Singh VK, Dwivedi P, Chaudhary BR et al (2015) Immunomodulatory effect of Gymnema syl-
vestre (R.Br.) leaf extract: an in vitro study in Rat model. PLoS One. https://doi.org/10.1371/
journal.pone.0139631
56. Costa JFO, David JPL, David JM et al (2008) Immunomodulatory activity of extracts
from Cordia superba Cham. and Cordia rufescens A. DC. (Boraginaceae), plant spe-
cies native from Brazilian semiarid. Rev Bras Farmaacogn. https://doi.org/10.1590/
S0102-695X2008000100004
57. Hussain A, Shadma W, Maksood A et al (2013) Protective effects of Picrorhiza kurroa on
cyclophosphamide-induced immunosuppression in mice. Protective effects of Picrorhiza
kurroa on cyclophosphamide-induced immunosuppression in mice. Pharmacogn Res. https://
doi.org/10.4103/0974-8490.105646
1322 S. P. Thekkekkoottumughath
58. Bafna A, Mishra S (2009) Antioxidant and immunomodulatory activity of the alkaloidal frac-
tion of Cissampelos pareira Linn. Sci Pharm. https://doi.org/10.3797/scipharm.0904-16
59. Dashputre NL, Naikwade NS (2010) Immunomodulatory activity of Abutilon indicum Linn.
on albino mice. IJPSR 1(3):178–184
60. Thakur M, Bhargava S, Dixit VK (2006) Immunomodulatory activity of Chlorophytum boriv-
ilianum Sant. F. Evid Based Complement Alternat Med. https://doi.org/10.1093/ecam/nel094
61. Bharshiv CK, Garg SK, Bhatia AK (2016) Immunomodulatory activity of aqueous extract
of Nyctanthes arbor-tristis flowers with particular reference to splenocytes proliferation and
cytokines induction. Indian J Pharmacol. https://doi.org/10.4103/0253-7613.186210
62. Sunil MA, Sunitha VS, Radhakrishnan EK et al (2019) Immunomodulatory activities of
Acacia catechu, a traditional thirst quencher of South India. J Ayurveda Integr Med. https://
doi.org/10.1016/j.jaim.2017.10.010
63. Ammon HPT (2010) Modulation of the immune system by Boswellia serrata extracts and
boswellic acids. Phytomedicine. https://doi.org/10.1016/j.phymed.2010.03.003
64. Mishra N, Tandon V, Gupta R (2012) Immunomodulation by Hibiscus rosa-sinensis: effect
on the humoral and cellular immune response of Mus musculus. Pak J Biol Sci. https://doi.
org/10.3923/pjbs.2012.277.283
65. Kori ML, Gaur M, Dixit VK (2009) Investigation of immunomodulatory potential of Cleome
gynandra Linn. Asian J Pharm Clin Res 2(1):35–39
66. Satpute KL, Jadhav MM, Karodi RS et al (2009) Immunomodulatory activity of fruits of
Randia dumetorum Lamk. JPP 1(3):36–40
67. Arreola R, Fabian SQ, Lopez-Roa RI et al (2015) Immunomodulation and anti-inflammatory
effects of garlic compounds. J Immunol Res. https://doi.org/10.1155/2015/401630
68. Manu KA, Kuttan G (2009) Immunomodulatory activities of Punarnavine, an alka-
loid from Boerhaavia diffusa. Immunopharmacol Immunotoxicol. https://doi.
org/10.1080/08923970802702036
69. Rahayu RP, Prasetyo RA, Purwanto DA et al (2018) The immunomodulatory effect of
green tea (Camellia sinensis) leaves extract on immunocompromised Wistar rats infected by
Candida albicans. Vet World. https://doi.org/10.14202/vetworld.2018.765-770
70. Friedman H, Klein TW, Newton C et al (1995) Marijuana, receptors and immunomodulation.
Adv Exp Med Biol. https://doi.org/10.1007/978-1-4615-1951-5_15
71. Punturee K, Wild CP, Kasinrerk W et al (2005) Immunomodulatory activities of Centella
asiatica and Rhinacanthus nasutus extracts. Asian Pac J Cancer Prev 6(3):396–400
72. Jayathirtha MG, Mishra SH (2004) Preliminary immunomodulatory activities of
methanol extracts of Eclipta alba and Centella asiatica. Phytomedicine. https://doi.
org/10.1078/0944711041495236
73. Srinivasan R (2019) Evaluation of immunomodulatory activity of the ethanolic extract of
Evolvulus alsinoides. Int J Pharmacol Toxicol Sci 9(2):32–38
74. Kshirsagar S, Thakur AS, Kshirsagar J (2015) Immunomodulatory and antioxidative prop-
erties of Clitoria ternatea. Int J Plant Sci. https://doi.org/10.15740/HAS/IJPS/10.2/158-162
75. Nfambi J, Bbosa GS, Sembajwe LF et al (2015) Immunomodulatory activity of methano-
lic leaf extract of Moringa oleifera in Wistar albino rats. J Basic Clin Physiol Pharmacol
26(6):603–611. https://doi.org/10.1515/jbcpp-2014-0104
76. Sunila ES, Kuttan G (2004) Immunomodulatory and antitumor activity of Piper longum Linn
and piperine. J Ethnopharmacol. https://doi.org/10.1016/j.jep.2003.10.016
77. Mallaiah GK, Raju PK, Thirupathi K et al (2016) Evaluation of immunomodulatory activity
of Tribulus terrestris in animal model. Int J Pharm Biol Sci 6(2):171–177
78. Davis L, Kuttan G (2000) Immunomodulatory activity of Withania somnifera. J
Ethnopharmacol. https://doi.org/10.1016/s0378-8741(99)00206-8
79. Ramprasath VR, Shanthi P, Sachdanandam P (2006) Immunomodulatory and anti-
inflammatory effects of Semecarpus anacardium LINN. Nut milk extract in experimental
inflammatory conditions. Biol Pharm Bull. https://doi.org/10.1248/bpb.29.693
Plants with Immunomodulatory Potential Described in Ayurveda 1323
100. Laxmi V (2015) Investigating the immunomodulatory effect of Cassia fistula on albino rats.
Adv Pharm Ethnomed. https://doi.org/10.14737/journal.ape/2015/3.1.1.5
101. Niphade SR, Asad M, Chandrakala GK et al (2009) Immunomodulatory activity of
Cinnamomum zeylanicum bark. Pharm Biol. https://doi.org/10.3109/13880200903019234
102. Soumaya KJ, Dhekra M, Fadwa CG et al (2013) Pharmacological, antioxidant, genotoxic
studies and modulation of rat splenocyte functions by Cyperus rotundus extracts. BMC
Complement Altern Med. https://doi.org/10.1186/1472-6882-13-28
103. Bouadi H, Necib Y, Bahi A (2015) Immunomodulatory activity of lectins extracted from
illicium Verum. Int J Pharm Sci Rev Res 31(1):129–131
104. Cosentino M, Bombelli R, Conti A et al (2009) Antioxidant properties and in vitro immuno-
modulatory effects of peppermint (Mentha x piperita l.) essential oils in human leukocytes. J
Pharm Sci Res 1(3):33–43
105. Mahamat O, Flora H, Tume C et al (2020) Immunomodulatory activity of momordica charan-
tia L. (Cucurbitaceae) leaf diethyl ether and methanol extracts on Salmonella typhi -infected
mice and LPS-induced phagocytic activities of macrophages and neutrophils. Evid Based
Complement Altern Med. https://doi.org/10.1155/2020/5248346
106. Eftekhar N, Moghimi A, Mohammadian RN et al (2019b) Immunomodulatory and anti-
inflammatory effects of hydroethanolic extract of Ocimum basilicum leaves and its effect on
lung pathological changes in an ovalbumin-induced rat model of asthma. BMC Complement
Altern Med. https://doi.org/10.1186/s12906-019-2765-4
107. Jantan I, Ilangkovan MY, Mohamad HF (2014) Correlation between the major components
of Phyllanthus amarus and Phyllanthus urinaria and their inhibitory effects on phagocytic
activity of human neutrophils. BMC Complement Altern Med. https://doi.org/10.1186/1472-
6882-14-429
108. Majdalawieh AF, Carr RI (2010) In vitro investigation of the potential immunomodulatory
and anti-cancer activities of black pepper (Piper nigrum) and cardamom (Elettaria cardamo-
mum). J Med Food. https://doi.org/10.1089/jmf.2009.1131
109. Muniandy K, Gothai S, Badran KMH et al (2018) Suppression of proinflammatory cyto-
kines and mediators in LPS-Induced RAW 264.7 macrophages by stem extract of alter-
nanthera sessilis via the inhibition of the NF-κB pathway. J Immunol Res. https://doi.
org/10.1155/2018/3430684
110. Labsi M, Khelifi L, Mezioug D et al (2016) Antihydatic and immunomodulatory effects of
Punica granatum peel aqueous extract in a murine model of echinococcosis. Asian Pac J Trop
Med. https://doi.org/10.1016/j.apjtm.2016.01.038
111. Gupta A, Chaphalkar SR (2016) Immunopharmacological screening of aqueous root extract
of Santalum album. J Herb Med Pharmacol 5(1):7–11
112. Chen HC, Chou CK, Lee SD et al (1995) Active compounds from Saussurea lappa Clarks that
suppress hepatitis B virus surface antigen gene expression in human hepatoma cells. Antivir
Res. https://doi.org/10.1016/0166-3542(94)00083-K
113. Bafna AR, MishraSH (2007) Immunomodulatory activity of petroleum ether extract of
flower heads of Sphaeranthus indicus linn. J Herb Pharmacother. https://doi.org/10.1300/
J157v07n01_03
114. Carrasco FR, Schmidt G, Romero AL et al (2009) Immunomodulatory activity of zingiber
officinale Roscoe, salvia officinalis L. and syzygium aromaticum L essential oils: evidence
for humor- and cell-mediated responses. J Pharm Pharmacol. https://doi.org/10.1211/
jpp/61.07.0017
115. Shivaprasad HN, Kharya MD, Rana AC et al (2006) Preliminary immunomodula-
tory activities of the aqueous extract of Terminalia chebula. Pharm Biol. https://doi.
org/10.1080/13880200500530542
116. Yu ZP, Xu DD, Lu LF et al (2016) Immunomodulatory effect of a formula developed from
American ginseng and Chinese jujube extracts in mice. J Zhejiang Univ Sci B. https://doi.
org/10.1631/jzus.B1500170
Plants with Immunomodulatory Potential Described in Ayurveda 1325
117. Chandan G, Kumar C, Verma K et al (2020) Datura stramonium essential oil composi-
tion and it’s immunostimulatory potential against colon cancer cells. 3 Biotech. https://doi.
org/10.1007/s13205-020-02438-4
118. Han X, Parker TL (2017) Cardamom (Elettaria cardamomum) essential oil significantly
inhibits vascular cell adhesion molecule 1 and impacts genomewide gene expression in
human dermal fibroblasts. Cogent Med. https://doi.org/10.1080/2331205x.2017.1308066
119. Uttara J, Mishra SH (2009) Preliminary evaluation of immunomodulatory and antistress
activity of methanol extract of Hedychium spicatum. Pharmacologyonline 1:1057–1071
120. Mishra A, Thakur M, Alok S (2016) Evaluation of immunomodulatory activity of polysac-
chride fraction of Inula racemosa, Bombax ceiba and Allium sativum. Int J Pharm Sci Res.
https://doi.org/10.13040/IJPSR.0975-8232
121. Daoudi A, Aarab L, Abdel-Sattar E (2013) Screening of immunomodulatory activity of total
and protein extracts of some Moroccan medicinal plants. Toxicol Ind Health. https://doi.
org/10.1177/0748233711430972
122. Rai SN, Birla H, Zahra W et al (2017) Immunomodulation of Parkinson’s disease using
Mucuna pruriens (Mp). J Chem Neuroanat. https://doi.org/10.1016/j.jchemneu.2017.06.005
123. Khorrami S, Daneshmandi S, Mosayebi G (2018) Sesame seeds essential oil and Sesamol
modulate the pro-inflammatory function of macrophages and dendritic cells and promote Th2
response. Med J Islam Repub Iran. https://doi.org/10.14196/mjiri.32.98
124. Sawant C, Joshi N, Reddy S et al (2014) Immunomodulatory medicinal plants of India: a
review. Int J Pharmacol Toxicol 4(2):109–115
125. Chulet R, Pradhan P (2010) A review on rasayana. Phcog Rev 3(6):229–e234
126. Wagner H (1984) In: Hikino H, Farnsworth NR (eds) Economic and medicinal plant research,
vol 1. Academic Press, London, pp 113–e153
Plant-Derived Drugs for Alzheimer’s
Disease and Other Neurological Disorders
1 Introduction
Dementia often begins with the gradual loss of intellectual functions of the brain,
such as memory, thinking, decision-making, learning capacity, attention, language,
and visuo-spatial functions. Dementia is not specific to any disease but is rather
caused by many neurological diseases including Alzheimer’s disease (AD) vascular
body dementia, Lewy body dementia, and Parkinson’s disease [1, 2]. Alzheimer’s
disease is a neurodegenerative disease that causes nerve cell death and is the world-
wide leading cause of dementia among 60–80% of patients. It is chronic, slowly
progressive with late onset majorly associated with elderly people over the age of
65. The early signs associated are dementia, cognitive decline, and anxiety that
decline the quality of daily living. Over time, these signs grow and become progres-
sively more severe affecting the person’s social and behavioral activities that lead to
depressive feelings and finally death [3–5]. According to Alzheimer’s Association,
50 million people globally are affected by Alzheimer’s disease (AD) which is
expected to triple by 2050 and increase to 150 million [6, 7]. Even the reported AD
and dementia-related deaths are increasing globally due to many factors including
growing population, aging, and increase in average lifetime. The death rate increased
by more than 145% between 2000 and 2019 in the US population alone, thus creat-
ing a huge socio-economic burden on the healthcare system and society at large
[3, 8, 9].
Withania somnifera
3.1
3.1.1 Neuroprotective Property
Ashwagandha has been demonstrated to have free radical scavenging and antioxi-
dant properties and also the ability to help maintain a robust immunity. Free radicals
created during the onset and course of Alzheimer’s disease have been demonstrated
to be scavenged by a subset of these components. Furthermore, these compounds
prevent amyloid-induced cell death in PC-12 cells and rat neural cells. Studies sug-
gest that prominent Withanamide A and Withanamide C [20] (Fig. 1a, b) extracted
from the fruits of somnifera plant protect PC-12 cells against amyloid peptide-
induced cell toxicity. Withanamides A and Withanamides C cohere to the β-amyloid
and suppress fibril formation [21].
3.1.2 Antioxidant Property
Various experiments were conducted on W. somnifera and the extract obtained from
it proved its effectiveness in the treatment of AD and its related neurological dys-
functions [22]. One such study reveals that WS extract shields SK-N-SH cells
1330 B. Sumithra et al.
Fig. 1 Major
Withanamides of Withania
somnifera: (a) structure of
Withanamide A (WA) and
(b) structure of
Withanamide C (WC)
(neuroblastoma cell line) from toxicity caused by Aβ peptide and acrolein and it was
also able to lower the levels of ROS which in turn reduces the oxidative stress in
SK-N-SH cells [23].
3.1.4 Neuro-regenerative Property
From this, we can demonstrate that the Withania somnifera extracts have promis-
ing pharmacological potential. It exhibits various properties, that is, neuroprotec-
tion, neuroregeneration, AChE inhibitory activity, and antioxidant activity, which
are key segments that have to be considered in producing a potential pharmacologi-
cal effect against Alzheimer’s activity.
3.1.5
Withania somnifera and Other Neurodegenerative Diseases
Centella asiatica is an annual herb of the Apiaceae family. It is one of the most
significant revitalizing flora for the cells of CNS in Ayurveda, and it is said to be
capable of boosting intellect, vitality, and memory. Triterpenes, asiaticoside, vella-
rin, asiatic acid, adecassoside, glycosides, madecassic acid, sapogenins, and centel-
loside are among the bioactive substances found in it [16]. Centella asiatica is
believed to enhance levels of the neurotrophic factor BDNF (brain-derived neuro-
trophic factor) by activating MAP kinases (mitogen-activated protein kinase) [25]
(Fig. 4). It is also renowned for its potential to enhance the healing of wounds in
addition to its cognitive boosting properties. Centella asiatica blocks enzymes that
disintegrate collagen while enhancing collagen production in preclinical trials,
resulting in a faster wound healing rate [26]. Various in vivo and in-vitro experi-
ments were conducted on gotu kola and numerous animal models were made to
know about the working mechanisms of various extracts of C. asiatica. Some of the
properties were thoroughly explained using animal models [27]. A few of them are
mentioned in Fig. 5.
3.2.1 Neuroprotective property
study. The pre-and post-colchicine Cornu Ammonis (CA) therapy boosted cogni-
tion, lowered malondialdehyde and nitrite concentrations, improved GSH levels,
and boosted glutathione-S-transferase, catalase, and SOD activities [31].
3.2.2 Antioxidant Property
Oxidative stress (OS) seems to be a key event in AD’s etiology. At the same time,
aggregated Ab-induced oxidative stress is a trigger in AD’s progression. Superoxide
is considered to be the main ROS in the human body and it can be produced by the
action of aggregated Ab1e40 on mitochondria which induces the activity of SOD
(superoxide dismutase). The extracts of CA have been shown to significantly reduce
this SOD activity and at the same time reverse the inhibition produced by SOD in
neural cells. The only drawback to this activity is that the extracts work in a dose-
dependent manner. These extracts also can moderate the activity of the enzyme
catalase which helps in the hydrolysis of peroxide (H2O2) into water and oxygen
based on cellular requirements. The data obtained from the experiment conducted
on mice model for Alzheimer’s disease have suggested that the extracts of Centella
might increase GR and GPx activity (antioxidant enzymes which help in the break-
down of ROS) in differentiated PC12 (for neurotoxicity) and IMR32 cells (neuro-
blastoma cell lines), reducing oxidative stress in these cells [32]. Glutathione is one
of the most common non-peptide intracellular thiols in cells, which is essential for
cellular oxidative metabolism. It would be assumed that raising glutathione levels
would lower ROS levels and oppose apoptotic signals [30].
Evodia rutaecarpa Benthem, one of the most common species used in primitive
Chinese medicine, is a fruit of the genus Evodia that is used to cure headaches,
stomach discomfort, postpartum bleeding, diarrhea, physiological alterations
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1335
Salvia officinalis
3.4
hyperglycemia are all the conditions wherein S. officinalis has often been used to
provide the right treatment in the domain of folk medicine. This plant has been the
subject of intensive research in the past few years, intending to document its classic
usage and identify undiscovered biological effects. It has been found to have several
biological effects such as anticancer [37], antinociceptive, antioxidant, anti-
inflammatory [38], antimicrobial [39], antidementia, and antimutagenic properties
so far [40].
The most important phytochemical components extracted from the various plant
parts such as leaves, roots, stems, and flowers include carbohydrates and fatty acids;
steroids; mono, di, tri and sesquiterpenoids and certain phenolic compounds,
namely, tannins and flavonoids [41–43]. The majority of these were extracted from
the aqueous extract, butanol fraction, alcoholic extract, or ethanoic extract of
the plant.
The severe loss of the functioning of cholinergic neurons is a key characteristic
of AD. Salvia officinalis has cholinergic properties; that is, it can mimic the action
of acetylcholine which is a neurotransmitter. In various research, it has, therefore,
been shown to have the potential to act as a source of innovative treatment for
AD. The research was conducted to assess the efficacy and the dependency of a
Salvia officinalis extract in individuals aged between 65 and 80 years with AD rang-
ing from a very mild to a serious condition of the disease for 4 months using a fixed
dosage of 60 drops/day. It was a placebo-controlled trial carried out in three differ-
ent centers in Iran. As a result of these, the extract from S. officinalis performed
better in terms of cognitive functions than the placebo-controlled group. No signifi-
cant variations in side effects between the two groups were noticed and in the S. offi-
cinalis extract group, the agitation of patients was overcome, which however
requires further confirmation. Thus, it was shown that a four-month therapy with a
hydroalcoholic extract enhanced cognitive functioning, according to Akhondzadeh
et al.’s randomized controlled experiment [44].
When looking at the mechanism, a possible interaction with the cholinergic sys-
tem has been hypothesized as one of the processes responsible for S. officinalis
cognitive and memory-enhancing effects. In research conducted on animals, the
ethanolic extract of the leaves used showed an increased memory retention capacity
in rats. Its interaction with nicotinic and muscarinic cholinergic systems was the
major factor responsible for the entire process of memory retention [45]. The essen-
tial oil (EO) from the S. officinalis leaf extract was found to have an intriguing
antioxidant activity with an IC50 value of 8.31 ± 0.55 mg/L. The inhibitory potency
of EO against the three enzymes 5-lipoxygenase (5-LOX), acetylcholinesterase
(AChE) and xanthine oxidase (XOD) was investigated for the first time in Tunisia.
The result was that the investigated enzymes AChE (IC50 = 38.71 ± 2.09 mg/L),
5-LOX (IC50 = 36.15 ± 1.27 mg/L) and XOD (IP (percent) = 36.89 ± 1.83 at a final
concentration of 50 mg/L in the well) were all completely inhibited by S. officinalis
EO [46].
The leaves were also shown to have fungistatic, virustatic, and antibacterial bio-
logical activities, among others. In healthy mice, the hydroalcoholic extract of
S. officinalis and rosmarinic acid (Fig. 7a), one of its main flavonoids, improved
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1337
Fig. 7 Chemical structures of potential compounds of Salvia officinalis: (a) rosmarinic acid and
(b) ursolic acid
Rehmannia glutinosa
3.5
The generation of H2O2 and other reactive oxygen species is linked to neurodegen-
erative diseases. Catalpol (Fig. 8), an iridoid glycoside found in the root of the
Rehmannia glutinosa plant, protects cells and mice from a variety of harmful stim-
uli [52]. Increased levels of reactive oxygen species (ROS) at the intracellular level
produce oxidative stress, which is thought to be a typical cause of the death of
neurons [53]. The balance between ROS formation and anti-oxidative activities may
be disrupted as a result of aging and many neurodegenerative diseases such as
Alzheimer’s disease and Parkinson’s disease [54].
Glutathione is a powerful antioxidant that protects cells from a variety of reactive
oxygen species (ROS). Several linked enzymes confer various tasks for glutathione
in live cells, in addition to the direct elimination of ROS by glutathione. In the
metabolism of glutathione and the defense against ROS, astrocytes in the brain play
a key role [55]. At the cellular level, astrocytes use two antioxidant mechanisms that
can rapidly inactivate H2O2 to limit the risk of radical generation from peroxides.
Reduced glutathione (GSH) acts as an electron donor in a glutathione
1338 B. Sumithra et al.
Gastrodia elata
3.6
Gastrodia elata (GE) Blume (Orchidaceae) has been utilized in eastern countries
for ages as a traditional herbal anticonvulsant. Vertigo, tetanus, and general paraly-
sis are all treated with GE as an analgesic and sedative. Some of the key ingredi-
ents are gastrodin, vanillin, and hydroxybenzaldehyde. Organic acids, glucose,
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1339
The dried bark of Cinnamomum cassia is known as Cinnamomi cortex (CC), and it
has been used to treat several potentially fatal ailments in traditional south Asian
medicine. Cinnamomi cortex possesses glucose and lipid metabolism improvement,
neuroprotection, anti-inflammatory, antioxidant, anticancer and other pharmaco-
logical properties [64].
At specific concentrations, three samples of cinnamon tannin demonstrate sig-
nificant scavenging capacity against hydroxyl and 2,2-diphenyl-1-picrylhydrazyl
(DPPH) radicals. The clearance rates of 2-diphenyl1-picrylhydrazyl radicals are
1340 B. Sumithra et al.
90.98%, 90.48%, and 89.38%, respectively, with the concentration of ethyl ethano-
ate extract, resin purification extract and crude extract about 0.04 mg/mL–1. In all
three samples, the hydroxyl radical clearance rates exceeded 65% and rose dose-
dependently range from 0.1 to 0.6 mg/mL–1. In the future, Cinnamomi cortex tannin
could be employed as a natural antioxidant [65].
The therapeutic efficacy of Cinnamomi cortex extract on neurodegenerative dis-
eases like Alzheimer’s disease (AD) induced Drosophila and transgenic mice has
been studied. According to the researchers, the lifetime of Drosophila induced with
AD was extended, the motor capability was restored and the harmful Ab oligomer
present in its central nervous system was destroyed. After ingesting the Cinnamomi
cortex extract, transgenic mice had fewer 56 kDa Ab oligomers and plaques, as well
as improved cognitive performance. According to the findings, CC extract protects
PC12 cells from toxicity by reducing the synthesis of hazardous Ab oligomers. CC
could be developed into an easy-to-use Alzheimer’s disease prevention and treat-
ment medicine. For the first time, Jana et al. discovered the mechanism through
which sodium benzoate; one of Cinnamomi cortex plant metabolite for neurodegen-
erative diseases [66].
When tested on mice, sodium benzoate raised neurotrophic factors derived from
the brain and neurotrophin-3 levels in the central nervous system via the PKA-
CREB pathway. These data show that CC could be utilized to treat neurodegenera-
tive illnesses as a main or secondary treatment. The Procyanidin type-a trimer
(trimer 1), (E)-3-phenylprop-2-enal and chromen-2-one extracted from Cinnamomi
cortex aqueous extract inhibited the swelling of the glial cell caused by deprivation
of glucose and oxygen, according to Panickar et al. [67]. Trimer 1 reduced nerve
cell swelling after ischemia injury by inhibiting the oxygen-free radical content and
circulation of calcium. It also reduced glutamate excitotoxicity by halting the reduc-
tion in glutamate uptake. The researchers concluded that CC could be utilized to
treat ischemia and other neurological issues [68].
4 Conclusions
For decades, numerous studies have been conducted to tackle different terrible neu-
rological conditions. Although a few medications are currently available for the
treatment of Alzheimer’s disease, synthetic drugs have several limitations. To date,
numerous drug candidates have been widely used in animal research and AD
patients, but no significant medication has been able to effectively cure the clinical
manifestations. Natural products, which involve the use of herbal drugs, could be a
viable cornerstone on which treatment procedures can be streamlined. It is undeni-
able that the demand for such medical therapy is growing. Medicinal plant-derived
biomolecules will help accelerate the development of new forms of treatment for
AD. Combining different plant remedies with potential neuroprotective effects as
MTDs could be useful in treating neurological disorders including Alzheimer’s dis-
ease. When these medications are used together, they start to improve drug efficacy
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1341
and patient adherence to therapy. In this chapter, numerous plants and various phy-
tochemicals and their potential applications for the treatment of neurodegenerative
disorders including Alzheimer’s disease are discussed.
It is also important to adopt a holistic approach that includes herbal medicinal
plants in our diet as well as trying to live in a collaborative environment that encour-
ages social, mental and spiritual activities, thereby reducing stress, which is a risk
factor for Alzheimer’s disease. Foreseeing the recent progress, we anticipate that the
intersection of medicinal plants will lead to successful therapeutic solutions for the
treatment of Alzheimer’s disease in near future.
References
1. Arvanitakis Z, Shah RC, Bennett DA (2019) Diagnosis and management of dementia: review.
JAMA 322(16):1589–1599. https://doi.org/10.1001/jama.2019.4782
2. https://www.who.int/news-room/fact-sheets/detail/dementia
3. Breijyeh Z, Karaman R (2020) Comprehensive review on Alzheimer’s disease: causes
and treatment. Molecules (Basel, Switzerland) 25(24):5789. https://doi.org/10.3390/
molecules25245789
4. Piau A, Nourhashémi F, Hein C, Caillaud C, Vellas B (2011) Progress in the development of
new drugs in Alzheimer;s disease. J Nutr Health Aging 15(1):45–57. https://doi.org/10.1007/
s12603-011-0012-x
5. Bateman RJ, Xiong C, Benzinger TL, Fagan AM, Goate A, Fox NC, Marcus DS, Cairns NJ,
Xie X, Blazey TM, Holtzman DM, Santacruz A, Buckles V, Oliver A, Moulder K, Aisen PS,
Ghetti B, Klunk WE, McDade E, Martins RN et al (2012) Clinical and biomarker changes
in dominantly inherited Alzheimer’s disease. N Engl J Med 367(9):795–804. https://doi.
org/10.1056/NEJMoa1202753
6. Yiannopoulou KG, Papageorgiou SG (2020) Current and future treatments in Alzheimer dis-
ease: an update. J Cent Nerv Syst Dis 12. https://doi.org/10.1177/1179573520907397
7. Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A,
Cohen-Mansfield J, Cooper C, Costafreda SG, Dias A, Fox N, Gitlin LN, Howard R, Kales
HC, Kivimäki M, Larson EB, Ogunniyi A, Orgeta V, Mukadam N (2020) Dementia preven-
tion, intervention, and care: 2020 report of the Lancet commission. Lancet (London, England)
396(10248):413–446. https://doi.org/10.1016/S0140-6736(20)30367-6
8. (2021) 2021 Alzheimer’s disease facts and figures. Alzheimers Dement 17(3):327–406. https://
doi.org/10.1002/alz.12328
9. GBD 2019 Dementia Forecasting Collaborators (2022) Estimation of the global prevalence
of dementia in 2019 and forecasted prevalence in 2050: an analysis for the Global Burden
of Disease Study 2019. Lancet Public Health 7(2):e105–e125. https://doi.org/10.1016/
S2468-2667(21)00249-8
10. Ayaz M, Ullah F, Sadiq A, Kim MO, Ali T (2019) Editorial: natural products-based drugs:
potential therapeutics against Alzheimer’s disease and other neurological disorders. Front
Pharmacol 10:1417. https://doi.org/10.3389/fphar.2019.01417
11. Ovais M, Zia N, Ahmad I, Khalil AT, Raza A, Ayaz M, Sadiq A, Ullah F, Shinwari ZK
(2018) Phyto-therapeutic and nanomedicinal approaches to cure Alzheimer’s disease: pres-
ent status and future opportunities. Front Aging Neurosci 10:284. https://doi.org/10.3389/
fnagi.2018.00284
12. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, Van der Flier
WM (2016) Alzheimer’s disease. Lancet (London, England) 388(10043):505–517. https://doi.
org/10.1016/S0140-6736(15)01124-1
1342 B. Sumithra et al.
13. DeTure MA, Dickson DW (2019) The neuropathological diagnosis of Alzheimer’s disease.
Mol Neurodegener 14(1):32. https://doi.org/10.1186/s13024-019-0333-5
14. Makhoba XH, Viegas C Jr, Mosa RA, Viegas FPD, Pooe OJ (2020) Potential impact of the
multi-target drug approach in the treatment of some complex diseases. Drug Des Devel Ther
14:3235–3249. https://doi.org/10.2147/DDDT.S257494
15. Shal B, Ding W, Ali H, Kim YS, Khan S (2018) Anti-neuroinflammatory potential of nat-
ural products in attenuation of Alzheimer’s disease. Front Pharmacol 9:548. https://doi.
org/10.3389/fphar.2018.00548
16. Rao RV, Descamps O, John V, Bredesen DE (2012) Ayurvedic medicinal plants for Alzheimer’s
disease: a review. Alzheimers Res Ther 4(3):22. https://doi.org/10.1186/alzrt125
17. Sehgal N, Gupta A, Valli RK, Joshi SD, Mills JT, Hamel E, Khanna P, Jain SC, Thakur
SS, Ravindranath V (2012) Withania somnifera reverses Alzheimer’s disease pathology by
enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci U S A
109(9):3510–3515. https://doi.org/10.1073/pnas.1112209109
18. Dar NJ, Ahmad M (2020) Neurodegenerative diseases and Withania somnifera (L.): an update.
J Ethnopharmacol 256:112769. https://doi.org/10.1016/j.jep.2020.112769
19. Halim MA, Rosli IM, Jaafar SSM, Ooi HM, Leong PW, Shamsuddin S, Najimudin N,
Azzam G (2020) Withania somnifera showed neuroprotective effect and increase longevity in
Drosophila Alzheimer’s disease model. BioRxiv. https://doi.org/10.1101/2020.04.27.063107
20. Kurapati KR, Atluri VS, Samikkannu T, Nair MP (2013) Ashwagandha (Withania som-
nifera) reverses β-amyloid1-42 induced toxicity in human neuronal cells: implications in
HIV-associated neurocognitive disorders (HAND). PLoS One 8(10):e77624. https://doi.
org/10.1371/journal.pone.0077624
21. Jayaprakasam B, Padmanabhan K, Nair MG (2010) Withanamides in Withania somnifera fruit
protect PC-12 cells from beta-amyloid responsible for Alzheimer’s disease. Phytother Res:
PTR 24(6):859–863. https://doi.org/10.1002/ptr.3033
22. Gregory J, Vengalasetti YV, Bredesen DE, Rao RV (2021) Neuroprotective herbs for the
management of Alzheimer’s disease. Biomol Ther 11(4):543. https://doi.org/10.3390/
biom11040543
23. Singh M, Ramassamy C (2017) In vitro screening of neuroprotective activity of Indian medici-
nal plant Withania somnifera. J Nutr Sci 6:e54. https://doi.org/10.1017/jns.2017.48
24. Konar A, Gupta R, Shukla RK, Maloney B, Khanna VK, Wadhwa R, Lahiri DK, Thakur MK
(2019) M1 muscarinic receptor is a key target of neuroprotection, neuroregeneration and
memory recovery by i-Extract from Withania somnifera. Sci Rep 9(1):13990. https://doi.
org/10.1038/s41598-019-48238-6
25. Chiroma SM, Baharuldin M, Mat Taib CN, Amom Z, Jagadeesan S, Ilham Adenan M, Mahdi
O, Moklas M (2019) Centella asiatica protects d-Galactose/AlCl3 mediated Alzheimer’s
disease-like Rats via PP2A/GSK-3β signaling pathway in their Hippocampus. Int J Mol Sci
20(8):1871. https://doi.org/10.3390/ijms20081871
26. Cai A, Xiao L, Zhou Y-P, Zhang Z-G, Yang Q-W (2020) Effect of Evodia rutaecarpa (Juss)
Benth extract on Alzheimer disease in mice. Trop J Pharm Res 19:823–828. https://doi.
org/10.4314/tjpr.v19i4.21
27. Soumyanath A, Zhong YP, Henson E, Wadsworth T, Bishop J, Gold BG, Quinn JF (2012)
Centella asiatica extract improves behavioral deficits in a mouse model of Alzheimer’s disease:
investigation of a possible mechanism of action. Int J Alzheimers Dis 2012:381974. https://
doi.org/10.1155/2012/381974
28. Dhanasekaran M, Holcomb LA, Hitt AR, Tharakan B, Porter JW, Young KA, Manyam BV
(2009) Centella asiatica extract selectively decreases amyloid beta levels in hippocampus of
Alzheimer’s disease animal model. Phytother Res: PTR 23(1):14–19. https://doi.org/10.1002/
ptr.2405
29. Centella asiatica (Gotu kola): cognitive vitality. https://www.alzdiscovery.org/cognitive-vital-
ity/ratings/centella-asiatica (Accessed on 15.02.2022)
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1343
30. Hafiz ZZ, Amin M, Johari James RM, Teh LK, Salleh MZ, Adenan MI (2020) Inhibitory
effects of raw-extract centella asiatica (RECA) on acetylcholinesterase, inflammations, and
oxidative stress activities via in vitro and in vivo. Molecules (Basel, Switzerland) 25(4):892.
https://doi.org/10.3390/molecules25040892
31. Kumar A, Dogra S, Prakash A (2009) Neuroprotective effects of Centella asiatica against
intracerebroventricular colchicine-induced cognitive impairment and oxidative stress. Int J
Alzheimers Dis 2009:972178. https://doi.org/10.4061/2009/972178
32. Chen CL, Tsai WH, Chen CJ, Pan TM (2015) Centella asiatica extract protects against amyloid
β1-40-induced neurotoxicity in neuronal cells by activating the antioxidative defence system. J
Tradit Complement Med 6(4):362–369. https://doi.org/10.1016/j.jtcme.2015.07.002
33. Fang Z, Tang Y, Ying J, Tang C, Wang Q (2020) Traditional Chinese medicine for anti-
Alzheimer’s disease: berberine and evodiamine from Evodia rutaecarpa. Chin Med 15:82.
https://doi.org/10.1186/s13020-020-00359-1
34. Cooper EL, Ma MJ (2017) Alzheimer disease: clues from traditional and complementary med-
icine. J Tradit Complement Med 7(4):380–385. https://doi.org/10.1016/j.jtcme.2016.12.003
35. Park CH, Kim SH, Choi W, Lee YJ, Kim JS, Kang SS, Suh YH (1996) Novel anticholines-
terase and antiamnesic activities of dehydroevodiamine, a constituent of Evodia rutaecarpa.
Planta Med 62(5):405–409. https://doi.org/10.1055/s-2006-957926
36. Zhang Y, Wang J, Wang C, Li Z, Liu X, Zhang J, Lu J, Wang D (2018) Pharmacological basis
for the use of evodiamine in Alzheimer’s disease: antioxidation and antiapoptosis. Int J Mol
Sci 19(5):1527. https://doi.org/10.3390/ijms19051527
37. Russo A, Formisano C, Rigano D, Senatore F, Delfine S, Cardile V, Rosselli S, Bruno M (2013)
Chemical composition and anticancer activity of essential oils of Mediterranean sage (Salvia
officinalis L.) grown in different environmental conditions. Food Chem Toxicol 55:42–47.
https://doi.org/10.1016/j.fct.2012.12.036
38. Wang M, Shao Y, Li J, Zhu N, Rangarajan M, LaVoie EJ, Ho CT (1999) Antioxidative phenolic
glycosides from sage (Salvia officinalis). J Nat Prod 62(3):454–456. https://doi.org/10.1021/
np980436g
39. Veličković DT, Ranđelović NV, Ristić MS, Veličković AS, Šmelcerović AA (2003) Chemical
constituents and antimicrobial activity of the ethanol extracts obtained from the flower, leaf
and stem of Salvia officinalis L. J Serb Chem Soc 68(1):17–24. https://doi.org/10.2298/
JSC0301017V
40. Ghorbani A, Esmaeilizadeh M (2017) Pharmacological properties of Salvia officinalis
and its components. J Tradit Complement Med 7(4):433–440. https://doi.org/10.1016/j.
jtcme.2016.12.014
41. Capek P, Hríbalová V (2004) Water-soluble polysaccharides from Salvia officinalis L. possess-
ing immunomodulatory activity. Phytochemistry 65(13):1983–1992. https://doi.org/10.1016/j.
phytochem.2004.05.020
42. Hayouni E, Chraief I, Abedrabba M, Bouix M, Leveau JY, Mohammed H, Hamdi M (2008)
Tunisian Salvia officinalis L. and Schinus molle L. essential oils: their chemical compositions
and their preservative effects against Salmonella inoculated in minced beef meat. Int J Food
Microbiol 125(3):242–251. https://doi.org/10.1016/j.ijfoodmicro.2008.04.005
43. Mitić-Ćulafić D, Vuković-Gačić BS, Knežević-Vukčević JB, Stanković S, Simić DM (2005)
Comparative study on the antibacterial activity of volatiles from sage (Salvia officinalis L.).
Arch Biol Sci 57(3):173–178. https://doi.org/10.2298/ABS0503173M
44. Akhondzadeh S, Noroozian M, Mohammadi M, Ohadinia S, Jamshidi AH, Khani M (2003)
Salvia officinalis extract in the treatment of patients with mild to moderate Alzheimer’s dis-
ease: a double blind, randomized and placebo-controlled trial. J Clin Pharm Ther 28(1):53–59.
https://doi.org/10.1046/j.1365-2710.2003.00463.x
45. Eidi M, Eidi A, Bahar M (2006) Effects of Salvia officinalis L (sage) leaves on memory reten-
tion and its interaction with the cholinergic system in rats. Nutrition (Burbank, Los Angeles
County, Calif.) 22(3):321–326. https://doi.org/10.1016/j.nut.2005.06.010
1344 B. Sumithra et al.
46. El Euch SK, Hassine DB, Cazaux S, Bouzouita N, Bouajila J (2019) Salvia officinalis essential
oil: chemical analysis and evaluation of anti-enzymatic and antioxidant bioactivities. S Afr J
Bot 120:253–260. https://doi.org/10.1016/j.sajb.2018.07.010
47. Hasanein P, Felehgari Z, Emamjomeh A (2016) Preventive effects of Salvia officinalis L. against
learning and memory deficit induced by diabetes in rats: possible hypoglycaemic and antioxi-
dant mechanisms. Neurosci Lett 622:72–77. https://doi.org/10.1016/j.neulet.2016.04.045
48. Porat Y, Abramowitz A, Gazit E (2006) Inhibition of amyloid fibril formation by polyphenols:
structural similarity and aromatic interactions as a common inhibition mechanism. Chem Biol
Drug Des 67(1):27–37. https://doi.org/10.1111/j.1747-0285.2005.00318.x
49. Chung YK, Heo HJ, Kim EK, Kim HK, Huh TL, Lim Y, Kim SK, Shin DH (2001) Inhibitory
effect of ursolic acid purified from Origanum majorana L on the acetylcholinesterase. Mol
Cells 11(2):137–143. PMID: 11355692
50. Lin HQ, Ho MT, Lau LS, Wong KK, Shaw PC, Wan DC (2008) Anti-acetylcholinesterase
activities of traditional Chinese medicine for treating Alzheimer’s disease. Chem Biol Interact
175(1–3):352–354. https://doi.org/10.1016/j.cbi.2008.05.030
51. Wu TY, Chen CP, Jinn TR (2011) Traditional Chinese medicines and Alzheimer’s disease.
Taiwan J Obstet Gynecol 50(2):131–135. https://doi.org/10.1016/j.tjog.2011.04.004
52. Jiang B, Shen RF, Bi J, Tian XS, Hinchliffe T, Xia Y (2015) Catalpol: a potential therapeutic
for neurodegenerative diseases. Curr Med Chem 22(10):1278–1291. https://doi.org/10.217
4/0929867322666150114151720
53. Choi K, Kim J, Kim GW, Choi C (2009) Oxidative stress-induced necrotic cell death via
mitochondira-dependent burst of reactive oxygen species. Curr Neurovasc Res 6(4):213–222.
https://doi.org/10.2174/156720209789630375
54. Liu Z, Zhou T, Ziegler AC, Dimitrion P, Zuo L (2017) Oxidative stress in neurodegenerative
diseases: from molecular mechanisms to clinical applications. Oxidative Med Cell Longev
2017:2525967. https://doi.org/10.1155/2017/2525967
55. Hossain MA, Piyatida P, da Silva JAT, Fujita M (2012) Molecular mechanism of heavy
metal toxicity and tolerance in plants: central role of glutathione in detoxification of reactive
oxygen species and methylglyoxal and in heavy metal chelation. J Bot 2012:37. https://doi.
org/10.1155/2012/872875
56. Forman HJ, Fukuto JM, Torres M (2004) Redox signaling: thiol chemistry defines which reac-
tive oxygen and nitrogen species can act as second messengers. Am J Physiol Cell Physiol
287(2):C246–C256. https://doi.org/10.1152/ajpcell.00516.2003
57. Farina C, Aloisi F, Meinl E (2007) Astrocytes are active players in cerebral innate immunity.
Trends Immunol 28(3):138–145. https://doi.org/10.1016/j.it.2007.01.005
58. Cacabelos R, Torrellas C, Carrera I, Cacabelos P, Corzo L, Fernández-Novoa L, Tellado I,
Carril JC, Aliev G (2016) Novel therapeutic strategies for dementia. CNS Neurol Disord Drug
Targets 15(2):141–241. https://doi.org/10.2174/1871527315666160202121548
59. Yang C, Shi Z, You L, Du Y, Ni J, Yan D (2020) Neuroprotective effect of catalpol via anti-
oxidative, anti-inflammatory, and anti-apoptotic mechanisms. Front Pharmacol 11:690. https://
doi.org/10.3389/fphar.2020.00690
60. Teoh ES (2016) Galeola to gymadenia. In: Medicinal orchids of Asia. Springer, Cham,
pp 375–440. https://doi.org/10.1007/978-3-319-24274-3
61. Jang JH, Son Y, Kang SS, Bae CS, Kim JC, Kim SH, Shin T, Moon C (2015)
Neuropharmacological potential of Gastrodia elata Blume and its components. Evid Based
Complement Alternat Med 2015:309261. https://doi.org/10.1155/2015/309261
62. Han YJ, Je JH, Kim SH, Ahn SM, Kim HN, Kim YR, Choi YW, Shin HK, Choi BT (2014)
Gastrodia elata shows neuroprotective effects via activation of PI3K signaling against oxidative
glutamate toxicity in HT22 cells. Am J Chin Med 42(4):1007–1019. https://doi.org/10.1142/
S0192415X14500633
63. Azam F (2010) Therapeutic potential of free radical scavengers in neurological disorders.
Handbook of free radicals: formation, types and effects. Nova Publishers, New York, pp 57–97
Plant-Derived Drugs for Alzheimer’s Disease and Other Neurological Disorders 1345
1 Introduction
Essential oils (EOs), secreted by plants, are primarily meant for various physiologi-
cal, ecological and defence activities. They belong to various classes, that is, iso-
lates of fatty acids, isoprenoids and phenolic complexes, and can be secreted by
various organs of plants. The EO market globally has been predicted to grow at a
compound annual growth rate (CAGR) of ~8–10% and will reach a staggering fig-
ure in excess of USD ten billion in the next 5 years or so. Essential oil contents in
different plant species are influenced by genetic and climatic conditions; the same
plant species show different constituents in different ecoclimatic areas. Plants from
tropical climates possess highly effective antimicrobial components that can be
A. N. Basha · K. Chithan
PG Department of Zoology, Arulmigu Palaniandavar College of Arts & Culture, Palani, Tamil
Nadu, India
R. Subramanian
The Thavaram Trust, Madurai, Tamil Nadu, India
G. G. Vincent · K. Murugesan · A. Ramachandran · S. Pethanan
Department of Zoology, Arumugam Pillai Seethai Ammal College, Tiruppathur, Tamil Nadu,
India
M. Panagal
Department of Biotechnology, Annai College of Arts and Science, Kumbakonam, Tamil
Nadu, India
C. P. Palanisamy (*)
State Key Laboratory of Biobased Materials and Green Paper Making, School of Food
Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences,
Jinan, Shandong, China
R. Jayakumararaj (*)
Department of Botany, Government Arts College, Madurai, Tamil Nadu, India
Lectins and peptides are positively charged biomolecules with disulphide bonds and
inhibit microbial growth. Both work upon microbial membranes and form ion chan-
nels competitively and inhibit microbial proteins and bind with polysaccharide
receptors [31]. Thionin, a peptide commonly isolated from barley and wheat, con-
tains 47 amino acid residues [39]. It is highly toxic to yeast and Gram-positive
bacteria [16]. Another peptide Febatin isolated from faba beans contains 47 amino
acid residues, structurally similar to thionin and effective against Escherichia coli,
Pseudomonas aeruginosa and Enterococcus hirae. Few plant species contain phen-
ylpropanoids as EO constituents, which are made up of a chain of three carbons
bound to an aromatic ring of six carbons and are mainly derived from phenylala-
nine [61].
1.2 Alkaloids
isolated from the plants of the buttercup family, have shown antimicrobial proper-
ties [44]. Solamargine, a glycoloid from the berries of Solanum khasianum, is useful
against HIV infection [38]. Some alkaloids have microbiocidal effects and are
active against Giardia, Entamoeba species and diarrhoea [20]. Berberine (an alka-
loid) is potentially used against trypanosomes [18].
1.3 Phenolic Compounds
1.4 Quinones
Quinones are highly reactive aromatic compounds with two ketone substitutions.
These are formed before the melanin synthesis pathway in the skin and are ubiqui-
tous. Quinones appear after roughing reaction during fruit/vegetable chopping/
1350 A. N. Basha et al.
Flavonoids occur as C6-C3 units linked to an aromatic ring. These are hydroxylated
phenolic substances, synthesized in plants in response to microbial infection.
Flavonoids are highly active against viruses (Fig. 2). These are phenolic structures,
which contain a carboxyl group. Flavonoids are formed after the addition of a
3-hydroxyl group. Glycyrrhizin, chrysin and swertifracheside are active against
HIV viruses [46]. Quercetin, a flavonoid compound, was effective against HSV-1,
Poliovirus-1 and Parainfluenza virus 3, while naringin, hesperetin and catechin
were less effective than quercetin. Galangin from Helichrysum aureointens was
effective against HSV-1 viruses and Gram-positive bacteria. Alpinumisoflavone is
effective against schistosomal infection. The mode of action of both flavones and
flavonoids is membrane disruption and enzymatic bindings, which depends on
hydroxylation due to the presence of hydroxyl groups on their rings [60].
1.6 Tannins
Tannin is a group of polymeric phenolic substances used for the tanning of leather
and the precipitation of gelatin. These possess astringent properties and are present
in wood, bark, leaves, fruits and roots. These are very strong antimicrobial agents.
Tannins act by penetration through the membrane, adhesion, competitive inhibition
of enzymes and binding to cellular envelops. Tannins inhibit the growth of larvae,
affect moulting in insects and are inhibitors of reverse transcriptase enzyme. There
are two categories of tannins, hydrolyzable (gallic acid) and condensed (proantho-
cyanidins) [21].
1.7 Coumarins
Coumarins are formed by the fusion of benzene and pyrene rings and show diverse
biological activity (anti-inflammatory, antithrombic and vasodilatory). Warfarin is
taken as an oral anticoagulant and used as a potential rodenticide. Coumarins also
show antiviral properties and are found effective against Candidiasis [19].
1.8 Terpenoids
Terpenoids are the most diversified group of secondary plant metabolites, derived
from a basic structure of five carbons (C5H8) commonly known as isoprenoid unit.
Terpenoids are classified on the basis of isoprenoid units found in the skeleton.
These are phenylpropanoids that interact with the cell membrane and accumulate in
the lipidic bilayer of bacteria [34] isolated diterpenoids from Sagittaria pygmaea.
Terpenoids or terpenes are active against bacteria, viruses and protozoa, while trit-
erpenoids like betulinic acid significantly inhibit the growth of the HIV virus. The
ethanol-soluble fractions of purple clove yield terpenoids, which show excellent
activity against Bacillus subtilis, Staphylococcus aureus and Gram-negative bacte-
ria as well as Candida albicans. Tricholorbdat-A, diterpenoid, directly inhibits
Helicobacter pylori activity [11].
Essential oils are highly potent phytochemicals, with high antimicrobial potential in
comparison to synthetic drugs as proved by low MIC values obtained in vitro.
Further, these natural products can stop genetic transformation in microbes. These
phytochemicals are active against more than one microbial strain while antibiotics
1352 A. N. Basha et al.
are not. These are either water or buffer soluble, show the least residual effect in the
body and do not show molecular catalysis and cross-reactivity [47]. These phyto-
chemicals possess mixed functional groups and are too complex in their structure;
therefore, microbes cannot develop resistance easily. EOs are multicomponent plant
products and contain different chemical groups such as pinene, camphor, limonene,
linalool, pyrogallol and chamaecynone [55].
All such components show diverse action mechanisms. EOs in eukaryotic cells
act as prooxidants, affect cell membrane permeability and inhibit the function of
cell organelles such as mitochondria. An important characteristic of EOs and their
components is their binding with lipids of bacterial cell membranes and mitochon-
drial cell structures making them more permeable to water [28]. Phenolic com-
pounds contain a hydroxyl group which acts as a transmembrane carrier of
monovalent cations and protons. This system works well for ionophore antibiotics
but not for aromatic compounds such as methanol. Phenols due to a selection of
high acidity and dislocated electron system release protons from the hydroxyl
group [59].
Undissociated covalent ions diffuse out, come across the cytoplasmic membrane
and dissociate to release proton in microbial cells, while terpenoids and phenylpro-
panoids interact with the bacterial cell membranes [13]. This activity may be due to
the hydrophobic nature of cyclic hydrocarbons which allows them to interact with
the bacterial cell membrane and occupy space between fatty acid chains and accu-
mulate in the lipid bilayer. This interaction leads to some important conformational
changes in the membrane structure, affecting their stability and resulting in the
expansion and formation of pore and fluidification of bacterial cells [22].
Thus, bacterial cells face leakage of ions across the cell membrane and lose
transmembrane ionic gradient very fast. Bacterial cells, however, restore the ionic
disturbances and counterbalance by using ionic pumps that check the cell death but
take a large portion of energy and make cell growth very slow [12]. Phenolic com-
ponents of EOs also interact with chemical groups of proteins and enzymes, and
most do so with protein through hydrogen bridges and ionic or hydrophobic interac-
tions while non-phenolic compounds interact with other functional groups [25].
Cinnamaldehyde, a natural constituent of Cinnamon, activates bacterial nucleic
acids and proteins by alkylation or formation of cross bridges. It also inhibits RNA,
DNA and protein synthesis in bacterial cells. It is also possible that EOs might
coagulate some cell constituents and cause denaturation by ionic release. The sulf-
hydryl groups in active components of EOs may interact more favourable against
the bacterial cells. Thus, garlic oil is more suitable than any other EO to kill both
Gram-negative and Gram-positive bacteria. A few chemical groups such as ketone,
aldehyde and acid may also interact in a better way with bacterial cells and their
components and show high antimicrobial activity but their major mode of action is
unknown [15].
Few oil components in combination with others might show synergistic effects
against microbes. Oxidized phenolic compounds were more toxic to bacterial cells.
Flavonoids lacking hydroxyl group very effectively disrupted the microbial mem-
brane. However, more hydroxylated phenolic compounds act on the membrane
Properties and Mechanism of Antimicrobial Agents from Plant-Derived Essential Oils 1353
surface due to the presence of the hydroxyl group. Flavonoids are highly effective
antimicrobial substances and inhibit the growth of a variety of microorganisms.
These form complexes with extracellular and soluble bacterial proteins and also
bind to bacterial cell walls. Catechins are flavonoids that inhibit the growth of Vibrio
cholerae, Streptococcus and Shigella. Flavonoids were also found inhibitory to a
number of viruses. The mechanism of action of terpenoids is not well understood
but it is believed to be involved in membrane disruption due to their lipophilic activ-
ity [50].
Capsaicin, a terpenoid, has a wide range of biological activities in humans affect-
ing the neural and digestive systems. Gram-positive bacteria possess a thick proteo-
glycan layer which does not respond to EOs. Gram-negative bacteria inhibit the
membrane disruption action of many EOs and show increased resistance against
drugs. Klebsiella pneumoniae was most resistant to many EOs but least resistant to
oregano Gram-negative K. pneumoniae possess an innate defence which prevents
entry of EO and does not allow access to the fragile inner membrane [23].
Essential oils from higher and aromatic plants have shown growth inhibitory poten-
tial against microbes due to the presence of certain secondary metabolites. People
in different parts of the world traditionally use EOs (Table 1) and their components
for various microbial infections related to skin, fever, gut and respiratory tract [26].
Clove, rosemary and lavender oils have shown strong antibacterial and antifungal
properties. Cinnamon oil possesses anti-diabetic and anti-inflammatory activity,
while lemon and peppermint show anticancer activity. EOs from Carum carvi,
Coriandrum sativum and Foeniculum vulgare effectively work against various
pathogenic bacteria [49].
A large number of EOs have been found effective against Gram-positive and
Gram-negative bacteria. Oil from Melaleuca alternifolia showed activity against
E. coli, S. aureus and skin pathogens. Few EOs and their major constituents were
found effective in the gaseous state against Haemophilus influenzae, Streptococcus
pneumoniae, S. pyrogens and Staphylococcus aureus [24]. Plant EOs (PEOs) have
been used traditionally for the eradication of respiratory tract infection and as ethi-
cal medicines by the tribals. These have also been used in inhalation therapy for
chronic bronchitis, sinusitis and respiratory tract mycoses. In aromatherapy, inhaled
EO vapours augment the output of respiratory tract fluid and maintain the ventila-
tion and drainage of the sinuses [9].
Essential oils also restore tracheal choking and reduce the intensity of asthma
attacks. EO from Mentha species shows antimicrobial and antioxidant activity [41],
due to the presence of free radical scavenging capacity (RSC). EOs from three spe-
cies M. aquatica, M. longifolia and M piperita exhibited strong activity against
E. coli strains. EOs from Hypericum scabrum and Hypericum scarboides showed
broad-spectrum antimicrobial activity against disease pathogens at a concentration
1354 A. N. Basha et al.
Table 1 (continued)
EOs from Extraction
S. No. Medicinal plants plant parts methods Microorganisms References
5. Mentha Aerial Hydrodistillation E. coli, [43]
aquatica L. parts Salmonella enteritidis,
Salmonella typhi,
Micrococcus flavus,
Staphylococcus aureus,
Staphylococcus
epidermis,
Bacillus subtilis
6. Mentha Aerial Hydrodistillation E. coli, [43]
longifolia L. parts Salmonella enteritidis,
Salmonella typhi,
Micrococcus flavus,
Staphylococcus aureus,
Staphylococcus
epidermis,
Bacillus subtilis
7. Mentha piperita Aerial Hydrodistillation E. coli, [43]
L. parts Salmonella enteritidis,
Salmonella typhi,
Micrococcus flavus,
Staphylococcus aureus,
Staphylococcus
epidermis,
Bacillus subtilis
8. Hypericum Stems, Hydrodistillation E. coli, [4]
scabrum L. flowers Bacillus brevis,
and leaves Bacillus cereus,
Streptococcus pyogenes,
Pseudomonas
aeruginosa,
Staphylococcus aureus,
Candida albicans
9. Hypericum Stems, Hydrodistillation E. coli, [4]
scabroides flowers Bacillus brevis,
Robson & and leaves Bacillus cereus,
Poulter. Streptococcus pyogenes,
Pseudomonas
aeruginosa,
Staphylococcus aureus,
Candida albicans
10. Hypericum Stems, Hydrodistillation E. coli, [4]
triquetrifolium flowers Bacillus brevis,
Turra. and leaves Bacillus cereus,
Streptococcus pyogenes,
Pseudomonas
aeruginosa,
Staphylococcus aureus,
Candida albicans
(continued)
1356 A. N. Basha et al.
Table 1 (continued)
EOs from Extraction
S. No. Medicinal plants plant parts methods Microorganisms References
11. Sida cordifolia Fresh Steam distillation Staphyllococus aureus, [36]
L. leaves Staphyllococus
epidermidis, Candida
guilliermondii and
Trichosporon inkin
12. Hedychium Fresh Hydrodistillation Staphylococcus aureus, [14]
venustum rhizomes Bacillus cereus,
B. subtilis,
Serratia marcescens,
Pseudomonas
fluorescens,
P. aeruginosa,
Klebsiella pneumoniae,
Proteus vulgaris,
Escherichia coli,
Salmonella typhi
13. Hedychium Fresh Hydrodistillation Staphylococcus aureus, [14]
spicatum var. rhizomes Bacillus cereus,
acuminatum B. subtilis,
Serratia marcescens,
Pseudomonas
fluorescens,
P. aeruginosa,
Klebsiella pneumoniae,
Proteus vulgaris,
Escherichia coli,
Salmonella typhi
14. Hedychium Fresh Hydrodistillation Staphylococcus aureus, [14]
coronarium rhizomes Bacillus cereus,
B. subtilis,
Serratia marcescens,
Pseudomonas
fluorescens,
P. aeruginosa,
Klebsiella pneumoniae,
Proteus vulgaris,
Escherichia coli,
Salmonella typhi
15 Hedychium Fresh Hydrodistillation Staphylococcus aureus, [14]
flavescens rhizomes Bacillus cereus,
B. subtilis,
Serratia marcescens,
Pseudomonas
fluorescens,
P. aeruginosa,
Klebsiella pneumoniae,
Proteus vulgaris,
Escherichia coli,
Salmonella typhi
Properties and Mechanism of Antimicrobial Agents from Plant-Derived Essential Oils 1357
of 80 μL/mL. EO from Lippia sidoides contained carvacrol and thymol which were
effective against oral pathogens [4, 43].
Oils of Lemon and lime leaves were effective against infectious bacterial strains.
PEOs from Sida cordifolia (Malvaceae) contain volatile constituents active against
S. aureus, S. epidermidis, C. guilliermondii and Trichosporon inkin. PEOs from
Sida cordifolia are commonly used for the treatment of rheumatism, inflammation,
asthma and nasal congestion and were found effective against S. aureus and S. epi-
dermiditis at 8% (v/v) dose. Sesame seed oil from Sesame radiatum and Sesamum
indicum is used for wound healing; seeds are used as the staple food by local popu-
lation in Nigeria. It is cultivated by local farmers for obtaining seed oil [36].
EOs from the rhizome of Hedychium species possess bactericidal and fungicidal
activity. Oil is also used for the treatment of stomach ailments and infections. Oil of
H. coronarium is used for the treatment of swelling and inflammation in tumour.
Oils from H. spicatum, H. coronarium, H. acuminatum, H. gardenarium and
H. aurantiacum possess antimicrobial activity. Rhizome oils from H. coronarium
and H. spicatum var. acuminatum were active against S. aureus (MTCC 96), Bacillus
cereus (MTCC 430), B. subtilis (MTCC 491), Serratia marcescens (MTCC-97),
Pseudomonas fluorescens (MTCC 103), Salmonella typhi (MTCC-733) and
Candida albicans (MTCC 227). Day jasmine, Cestrum diurnum (Solanaceae),
Citrus nastudaidai and Hayata peel oil were highly effective against pathogenic
bacteria and proved to be strong antimicrobial agents [14].
Eugenol, a well-characterized oil constituent found in clove oil, was highly bac-
teriostatic and fungistatic. Olive oil shows bactericidal activity against harmful
intestinal microbiota (Clostridium perfringens and E. coli); it is also active against
beneficial microorganisms, Lactobacillus and Bifidobacterium bifidum. The antimi-
crobial compounds in olive oil are hydroxytyrosol, vanillic acid, p-coumaric acid,
ferulic acid and vanillin, which are highly effective against Listeria monocytogenes,
S. aureus, Salmonella enterica and Yersinia species. Juniper berry oil possesses
broad-spectrum pharmacological activities against bacterial and fungal strains. Its
strongest fungicidal activity was recorded against Candida species. EOs from the
leaves of Toddalia asiatica, Solidago microglossa and Cassia alata have shown
inhibitory activity against Gram-negative and Gram-positive bacteria [42].
Garlic is traditionally used as a natural medicine for the treatment of various
diseases. Its oil and powder were effective against bacterial pathogens by virtue of
systemic distribution in the small intestine. Garlic and its compounds are safer than
antibiotics in eradicating the infection caused by Helicobacter pylori. Aqueous gar-
lic extracts with omeprazole showed a synergistic effect against Helicobacter pylori.
Most of the commercial garlic preparations, available in the market, contain garlic
oil (GO) and allicin. Allicin derivatives allyl and methyl sulphide showed very high
antimicrobial potential and were effective against stomach cancer. Its diallyl com-
pounds were effective against H. pylori. Chile peppers contain provitamins A, E and
several B and are used in flavouring food materials. Its constituent capsaicin (terpe-
noid) showed biological activity against disease-causing pathogens in humans. It is
used as an analgesic and shows bactericidal activity against H. pylori [29].
1358 A. N. Basha et al.
Viral epidemic is a global concern, mandating an urgent need for antivirals. Some
viral diseases can be cured by antiviral drugs, but others still don’t have any vac-
cines/drugs. Most of the approved antiviral drugs are associated with side effects.
This eventually drives the need for the development of plant-based antivirals. Plants
contain a variety of phytochemicals like flavonoids, terpenoids, lignins, alkaloids
1360 A. N. Basha et al.
and coumarins that possess antioxidant activities and help inhibit the viral genome.
Various plant-derived products have been well studied against viruses like herpes
virus, human immunodeficiency virus (HIV), influenza and hepatitis virus. More
recently, Coronavirus disease (COVID-19) caused by a newly identified coronavi-
rus has become pandemic and affected the world’s population severely. However,
there are still less explored phytochemicals for the inhibition of HIV that causes
AIDS. In this chapter, an extensive investigation has been made on the applications
of EOs to control opportunistic infections (OIs) that occur due to bacterial and fun-
gal pathogens in HIV/AIDS patients. In recent years, there has been an increasing
interest in the use of plant-based natural products due to aspects concerning the
safety of synthetic compounds.
In addition to inducing resistance, antibiotics are sometimes associated with
opposing effects such as hypersensitivity, immune suppression and allergic reac-
tions. Therefore, there is a need to develop alternative antimicrobial drugs for the
treatment of OIs. At this point in time, investigations on plants used in traditional
medicines as potential sources of novel antimicrobial compounds gain due
prominence.
References
1. Assarch MH, Jaimand K, Rezaee MB (2007) Chemical composition of the essential oils of six
Eucalyptus species (Myrtacea) from South West of Iran. J Essent Oil Res 6:469–473
2. Bakhtiarizade M, Souri MK (2019) Beneficial effects of rosemary, thyme and tarragon essen-
tial oils on postharvest decay of Valencia oranges. Chem Biol Technol Agric 6:9
3. Begum J, Bhuiyan MNI, Chowdhury JU, Hoque MN, Anwar MN (2008) Antimicrobial activ-
ity of essential oil from seeds of Carum carvi and its composition. Bangladesh J Microbiol
25(2):85–89
4. Botelho MA, Nogueira NA, Bastos GM, Fonseca SG, Lemos TL, Matos FJ, Montenegro D,
Heukelbach J, Rao VS, Brito GA (2007) Antimicrobial activity of the essential oil from Lippia
sidoides, carvacrol and thymol against oral pathogens. Braz J Med Biol Res 40(3):349–356
5. Burt SA, Vlielander R, Haagsman HP, Veldhuizen EJ (2005) Increase in activity of essential
oil components carvacrol and thymol against Escherichia coli O157: H7 by addition of food
stabilizers. J Food Prot 68:919–926
6. Cafarchia C, De-Laurentis N, Milillo MA, Losacco V, Puccini V (2002) Antifungal activ-
ity of essential oils from leaves and flower of Inula viscosa (Asteraceae) by Apulian region.
Parasitologia 44:153–156
7. Calsam Ouattra B, Simard RE, Holley RA, Pictte GJP, Begin A (1997) Antibacterial activ-
ity of selected fatty acids and essential oil against six meat spoilage organism. Int J Food
Microorganism 37:155–162
8. Calsamiglia S, Busquet M, Cardozo PW, Castillejos L, Ferret A (2007) Invited review: essen-
tial oils as modifiers of rumen microbial fermentation. J Dairy Sci 90(6):2580–2585
9. Carson CF, Mee BJ, Riley TV (2002) Mechanism of action of Melaleuca alternifolia (tea tree)
oil on Staphylococcus aureus determined by time-kill, lysis, leakage, and salt tolerance and
electron microscopy. AAC 46(6):1914–1920
10. Cosentino S, Barra A, Pisano B, Cabizza M, Pirisi FM, Palmas F (2003) Composition and
antimicrobial properties of Sardinian Juniperus essential oils against food borne pathogens and
spoilage microorganisms. J Food Prot 66(7):1288–1291
Properties and Mechanism of Antimicrobial Agents from Plant-Derived Essential Oils 1361
11. Cowan MM (1999) Plant products as antimicrobial agents. Clin Microbiol Rev 12:564–582
12. Cox SD, Mann CM, Markam JL (2001) Interaction between components of the essential oil of
Melaleuca alternifolia. J Appl Microbiol 91:492–497
13. Dorman HJ, Deans SG (2000) Antimicrobial agents from plants: antibacterial activity of plant
volatile oils. J Appl Microbiol 88(2):308–316
14. Faleiro ML, Miguel MG, Ladeiro F, Vanancio F, Tavarses R, Brito JC, Figueiredo AC, Barrosa
JG, Pedro LG (2003) Antimicrobial activity of essential oils isolated from Portuguese endemic
species of Thymus. Lett Appl Microbiol 36:35–40
15. Feldberg RS, Chang SC, Kotik AN, Nadler M, Neuwirth Z, Sundstrom DC, Thompson NH
(1988) In vitro mechanism of inhibition of bacterial cell growth by allicin. Antimicrob Agent
Chemother 32(12):1763–1768
16. Fernandez MA, Garcia MD, Saenz MT (1996) Antibacterial activity of the phenolic acids frac-
tions of Scrophularia frutescens and Scrophularia sambucifolia. J Ethnopharmacol 53(1):11–14
17. Fontenelle ROS, Morias SM, Brito EHS, Kerntopt MR, Brilhante RSN, Cordeiro RA, Tome
AR, Queiroz MGR, Nascimento NRF, Sidrim JJC, Rocha MEG (2007) Chemical composi-
tion, toxicological aspects and antifungal activity of essential oil from Lippia sidoides Cham.
J Antimicrob Chemother 59:934–940
18. Freiburghaus F, Kaminsky R, Nkunya MHH, Brun R (1996) Evaluation of African medicinal
trypanocidal activity. J Ethano Pharmacol 55:1–11
19. Garg SS, Gupta J, Sharma S, Sahu D (2020) An insight into the therapeutic applications of
coumarin compounds and their mechanisms of action. Eur J Pharm Sci 152:105424
20. Ghoshal S, Krishna-Prasad BN, Lakshmi V (1996) Antiamoebic activity of Piper longum fruits
against Entamoeba histolytica in vitro and in vivo. J Ethanopharmacol 50:167–170
21. Girard M, Bee G (2020) Invited review: Tannins as a potential alternative to antibiotics to
prevent coliform diarrhea in weaned pigs. Animal 14(1):95–107
22. Griffin SG, Wyllie SG, Markham JL, Leach DN (1999) The role of structure and molecu-
lar properties of terpenoids in determining their antimicrobial activity. Flavour Fragrance J
14(5):322–332
23. Gustafson E, Cox SD, Liew SY, Wyllie SG, John R, Warmington J (2001) The bacterial mul-
tiple antibiotic resistant (Mar) phenotype leads to increased tolerance to tea tree oil. Pathology
33(2):211–215
24. Inouye S, Takizawa T, Yamaguchi H (2001) Antibacterial activity of essential oils and their
major constituents against respiratory tract pathogens by gaseous contact. J Antimicrob
Chemotherap 47(5):565–573
25. Juven BJ, Kanner J, Schved F, Weisslowicz H (1994) Factors that interact with the antibacterial
action of thyme essential oil and its active constituents. J Appl Bacteriol 76:626–631
26. Karaman S, Digrak M, Ravid U, Ilcim A (2001) Antibacterial and antifungal activity of the
essential oils of Thymus revolutus Celak from Turkey. J Ethnopharmacol 76(2):183–186
27. Kizil G, Toker Z, Ozen HC, Aytekin C (2004) The antimicrobial activity of essential oils of
Hypericum scabrum, Hypericum scabroides and Hypericum triquetrifolium. Phytother Res
18(4):339–341
28. Knobloch K, Weigand H, Weis N, Schwar HM, Vigenschow H (1986) Action of terpenoids
on energy metabolism. In: Progress in essential oil research: 16th international symposium on
essential oils. De Gruyter, Berlin, pp 429–445
29. Koch HP (1996) Biopharmaceutics of garlic’s effective compounds. In: Koch HP, Lawson P,
Lawson LD (eds) Garlic the science and therapeutic application of Allivum sativum. Williams
and Wilkins, Baltimore, pp 213–220
30. Kumar N, Goe N (2019) Phenolic acids: natural versatile molecules with promising therapeu-
tic applications. Biotechnol Rep (Amst) 24:e00370
31. Kurpe SR, Grishin SY, Surin AK, Panfilov AV, Slizen MV, Chowdhury SD, Galzitskaya OV
(2020) Antimicrobial and amyloidogenic activity of peptides. Can antimicrobial peptides be
used against SARS-CoV-2? Int J Mol Sci 21(24):9552
32. Lawless J (1995) The illustrated encyclopodia of essential oils. UK Elements Book Ltd,
Shafesbury, pp 1–47
1362 A. N. Basha et al.
33. Lee SB, Cha KH, Kim SN, Altantsetseg S, Shatar S, Sarangerel O, Nho CH (2007) The antimi-
crobial activity of essential oil from Dracocephalum foetidum against pathogenic microorgan-
isms. Microbiol 45(1):53–57
34. Liu XT, Pan Q, Shi Y, Williams ID, Sung HH, Zhang Q, Liang JY, Ip NY, Min ZD (2006)
Entrosane and labdane diterpenoides from Sagittaria and their antibacterial activity against
three oral pathogens. J Nat Prod 69(2):255–260
35. Lopez P, Sanchez C, Batlle R, Nerin C (2005) Solid and vapor-phase antimicrobial activities
of essential oils: susceptibility of selected food borne bacterial and fungal strains. J Agric Food
Chem 53(17):6939–6946
36. Lota M, de Rocca Serra D, Tomi F, Jacquemond C, Casanova J (2001) Volatiles components of
peel and leaf oils of lemon and lime species. Biochem Syst Ecol 29:77–104
37. Ma CM, Nakamura N, Hattori M (2001) Inhibitroy effect on HIV-1 protease of tri-
pcoumaroylspermidine from Artemesia caruifolia and amides. Chem Pharm Bull (Yokyo)
49:915–917
38. McMohan JB, Currens MJ, Gulakowki RJ, Buckheit RWJ, Lackman SC, Michellamine B
(1995) A novel plant alkaloid inhibits human immuno deficiency virus-induce distinct mecha-
nisms. Antimicrob Agents Chemother 39:484–488
39. Méndez J, Blanco L, Lazaro JM, Salas M (1994) Primer-terminus stabilization at the psi 29
DNA polymerase active site mutational analysis of conserved motif TX2GR. J Biol Chem
269(47):30030–30038
40. Mimica-Dukić N, Božin B, Soković M, Mihajlović B, Matavulj M (2003) Antimicrobial and
antioxidant activities of three Mentha species essential oils. Planta Med 69(05):413–419
41. Mimica-Dukic N, Bozin B, Sokovic M, Simin N (2004) Antimicrobial and antioxidant activi-
ties of Melissa officinalis L (Lamiaceae) essential oil. J Agri Food Chem 52(9):2485–2489
42. Morel AF, Dias GO, Poro C, Simionatto E, Stuker CZ, Dalcol II (2006) Antimicrobial activity
of extractives of Solidago microglossa. Fitoterapia 77(6):453–458
43. Oliveira DR, Leitao GG, Santos SS, Bizzo HR, Lopes D, Alviano CS, Alviano DS, Leitao
SG (2006) Ethnopharmacological study of two Lippia species from Oriximiná, Brazil. J
Ethnopharmacol 108(1):103–108
44. Omulokoli E, Khan B, Chhabra SC (1997) Antiplasmodial activity of four Kenyan medicinal
plants. J Ethanopharmacol 56:133–137
45. Othman L, Sleiman A, Abdel-Massih RM (2019) Antimicrobial activity of polyphenols and
alkaloids in middle eastern plants. Front Microbiol 10:911
46. Parhi B, Bharatiya D, Swain SK (2020) Application of quercetin flavonoid based hybrid nano-
composites: a review. Saudi Pharm J 28(12):1719–1732
47. Patra AK (2012) An overview of antimicrobial properties of different classes of phytochemi-
cals. Diet Phytochem Microb:1–32
48. Pepeljnjak S, Kosalec I, Kalodera Z, Blazevic N (2005) Antimicrobial activity of Juniper berry
essential oil (Juniperus communis L, Cupressaceae). Acta Pharma 55:417–422
49. Prabuseenivasan S, Jayakumar M, Ignacimuthu S (2006) In vitro antibacterial activity of some
plant essential oils. BMC Comp Altern Med 6(1):39–45
50. Reuter HD, Koch JP, Lawson L (1996) Therapeutic effects and application of garlic and its
preparations. In: Koch HP, Lawson LD (eds) Garlic: the science and therapeutic application of
Allium sativum L and related species. William Wilkins, Baltimore, pp 135–212
51. Reynolds JEF (1996) Martindale- the extra pharmacopoeia 31st Ed. Royal Pharmaceautical
Society of Great Britian, London
52. Saddi M, Sanna CA, Chisu F, Casu L, Bonsignore L, De-Logu A (2007) Antiherpevirus activ-
ity of Artemisia arborescens essential oil and inhibiton of lateral diffusion in Vero cells. Ann
Clin Micro Antimicrob 6:10–16
53. Saini R (2011) Coenzyme Q10: the essential nutrient. J Pharm Bioallied Sci 3(3):466–467
54. Silva F, Ferreira S, Queiroz JA, Domingues FC (2011) Coriander (Coriandrum sativum L.)
essential oil: its antibacterial activity and mode of action evaluated by flow cytometry. J Med
Microbiol 60:1479–1486
Properties and Mechanism of Antimicrobial Agents from Plant-Derived Essential Oils 1363
55. Soković M, Glamočlija J, Marin PD, Brkić D, Griensven LJLD (2010) Antibacterial effects of
the essential oils of commonly consumed medicinal herbs using an in vitro model. Molecules
15(11):7532–7546
56. Sourmaghi MHS, Amin G, Samadi N, Hemmati F, Sarkhail P (2006) Chemical composition
and antimicrobial activity of essential oil of Salvia spinosa L. Asian J Plant Sci 5(4):654–656
57. Sugumaran M (2016) Reactivities of Quinone Methides versus o-Quinones in Catecholamine
metabolism and Eumelanin biosynthesis. Int J Mol Sci 17(9):1576
58. Tellez MR, Dayan FE, Schrader KK, Wedge DE, Duke SO (2000) Composition of some
biological activities of the essential oils of Callicarpa americana. J Agric Food Chem
48(7):3008–3012
59. Ultee A, Bennik MH, Moezelaar RJ (2002) The phenolic hydroxyl group of carvacrol is essen-
tial for action against the food-borne pathogen Bacillus cereus. AEM 68(4):1561–1568
60. Upadhyay RK (2010) Essential oils: anti-microbial, antihelminthic, antiviral, anticancer and
anti-insect properties. J Appl Biosci 36(1):1–22
61. Upadhyay RK (2011) Plant natural products: their pharmaceutical potential against disease
and drug resistant microbial pathogens. J Pharm Res 4(4):1179–1185
Nanotechnological Modus Operandi
for the Delivery of Cytotoxic
Phytochemicals
1 Introduction
Cancer is becoming a major health concern day by day and is spreading both in
developing as well-developed countries. The emergence of drug resistance to cyto-
toxic agents has made it difficult to control the spread of this disease [1]. Apart from
chemotherapeutical approaches, the management of cancer involves radiation ther-
apy and surgical interventions. The currently available chemotherapeutic drugs for
cancer treatment include anti-tubulin drugs like taxanes, numerous molecular tar-
geting drugs, DNA interactive drugs such as doxorubicin (Fig. 1) and antimetabo-
lites like methotrexate [2]. The utmost drawback of these cytotoxic agents is the
emergence of resistance and variable toxic effects in patients. Traditional Indian and
Chinese medicine has listed out various medicinal plants that have anti-cancer
action and are being evaluated alongside the modern medicine system. Side effects
that occur from the usage of synthetic chemotherapeutic agents such as hair loss,
lowering of the immune response, increased susceptibility to opportunistic infec-
tions, cardiac toxicity, neurological and gastrointestinal disturbances can be mini-
mized by direct delivery of plant-based cytotoxic agents at the tumour site [2].
Various phytochemicals from plant origin (Table 1) have proved to reduce cell pro-
liferation, slow down the rate of metastases, induction of apoptosis and inhibition of
angiogenesis [10, 11]. Paclitaxel (Fig. 1), a taxane diterpene, is extracted from
Taxus brevifolia Nutt. and is actively used in the treatment of a variety of cancers
[12]. The site-specificity can be enhanced by site targeting as well as various side
effects and poor drug delivery can be reduced by appointing nanotechnological drug
delivery approaches. The conventional drug delivery approaches do not render the
chemotherapeutic agent to distinguish between normal and cancerous cells, nano-
technology engineers the drug to only interact and bind with cancer cells, thus
reducing the chances of organ damage [13]. Using nanotechnology, the cancer cells
can be actively or passively targeted. In active targeting, a targeting agent is attached
to the nanoparticle and it works via antibody/antigen or ligand/receptor interaction
[14, 15]. The vectors that are used for active targeting include synthetic, natural and
lipid polymers. For enhanced half-life, the nanomaterial is coated with hydrophilic
polymer to prevent washout and increased time in the bloodstream. The coating
Fig. 1 (continued)
even repels the plasma proteins leading to the prevention of opsonized [16]. The
most common hydrophilic polymers that are employed in cancer targeting include
poloxamine, polyethylene glycol, polysaccharide and poloxamer [17]. Passive tar-
geting of the cancer cells can be through the leaky blood vessels in the endothelial
cells, the delivery system can pass through the formed pores which are usually in
the range of 100–780 nm [18]. The lymphatic drainage is also faulty at the tumour
site, helping in the retention and accumulation of nanoparticles, which prolong the
drug action [19]. Nanotechnology has already transformed cancer treatment in
numerous ways and is fundamentally altering the treatment plan. It has had a sig-
nificant influence on selective cancer cell recognition, targeted drug administration
and overcoming the limits of traditional chemotherapies. The properties of
1368 T. Alex et al.
nanoparticles are due to their specific shape, surface characteristic features and size.
The nanoparticles which range in the size of 10–100 nm are mostly considered in
cancer treatment and can effectively deliver cytotoxic agents as well as achieve the
enhanced permeability and retention effect. Nanoparticles that are in the size range
of 100 nm can be easily targeted by the phagocytes and the ones which range
between 1 and 2 nm can be easily cleared out by the kidneys. The nanoparticles
based on their composition can be divided into organic nanoparticles (Polymer,
Liposome-based nanoparticles, Dendrimers), inorganic nanoparticles (Gold, Silver,
Silica, Magnetic nanoparticles, Carbon nanotubes, Quantum dots) and hybrid
nanoparticles (Cell membrane coated nanoparticle, Organic-inorganic, Lipid-
polymer nanoparticle) [20].
Efforts are being put forward to synthesize nanocarriers using eco-friendly tech-
niques to produce nontoxic and environmentally safe biomaterial. The nanoparticles
synthesized using green nanotechnology have good stability with suitable dimen-
sions [21]. The conventional synthesis method using chemical and physical meth-
ods has led to generation of toxic by-products which are environmentally hazardous.
The yield might be greater with conventional techniques but these methods are cost-
lier and complicated [22]. Green synthesis involves production of nanoparticles
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1369
using plants, microbes and their by-products such as lipids and proteins. This tech-
nique uses a bottom-up approach which uses a stabilizing and reducing agent and
has three main steps: choosing an appropriate solvent medium, choosing a reducing
agent which is environmentally benign and eco-friendly and the last step is use of
nontoxic capping agent for a stabilized nanoparticle [23]. The magnetic nanoparti-
cles are synthesized by using a nanoemulsion mixture consisting of sodium hydrox-
ide and iron sucrose. In a study conducted by Kuang et al. used green synthesis
technique to produce magnetic nanoparticles, utilizing the extracts from oolong,
black and green tea [24]. Tripathi et al. synthesized carbon nanotubes using green
leaves extract from rose, Kaner, walnut, neem and garden grass [25]. Alippilakkotte
et al. synthesized polylactic acid/silver nanofibres using extracts from the fruit of
Momordica charantia [26].
In this chapter, we would be highlighting various nanoparticles that have been
studied and thus, can be employed to deliver cytotoxic phytochemicals in different
types of cancers.
2 Liposome
The size of liposomes ranges between 10 nm and 2500 nm and they are made up of
phospholipids. The final shape of liposomes depends upon the molar concentration,
ion concentration of the medium and temperature of the medium [27]. On the basis
of size, liposomes can be classified into multi-lamellar (1.5 um), oligo-lamellar
(1–1.5 um), uni-lamellar (100–250 nm) and small uni-lamellar (20–100 nm). The
main benefit of liposomal formulation is the sustained drug release pattern which
lowers down the requirement of dependency on injectable drugs. Moreover, the
half-life of the drug is also scaled up and it remains in the body for longer time,
when given in the form of liposomes [28, 29]. Liposomes can be administered by
oral route as well as intravenous route, for local administrations, liposomes are
injected via intraperitoneal, subcutaneous, intramuscular and intradermal route
[30]. The mechanism of action of liposomes on cancer cells is their ability to accu-
mulate at the tumour site after which they are up taken by cancer cells and release
free drug [31]. The anticancer effect of curcumin (Fig. 1), which is derived from
Curcuma longa, is being tested out on a variety of cancers such as prostate, liver and
breast cancers. This phytochemical works by interfering with the translation of
Bcl-xl protein and in regulation of apoptosis by having an effect on the release of
cytochrome and reactive oxygen species as well as affecting the cell cycle by regu-
lation of molecular factors like cyclin [32]. Forming liposomes of curcumin will
lead to improvement in the pharmacodynamic and pharmacokinetic profile of this
drug, reduction in dose required to show anti-cancer effect as compared with free
curcumin [33]. Another well-established anticancer agent, paclitaxel, which is
extracted from Taxus brevifolia, is utilized in various cancers such as gastric, breast,
ovarian and non-small cell lung cancer [34, 35]. It leads to cell death in cancer cells
by enhancing the polymerization of protein tubulin during the cell division, further
1370 T. Alex et al.
stabilizing the microtubules, causing disruption in the stability and finally causing
cell death [36]. Its cytotoxic action reduces due to its poor water solubility and high
lipophilicity; when non-aqueous base vehicle is added, the formulation causes
hypertensive reactions. Paclitaxel liposomes having mitochondrial targeting action
can be formulated by mixing cholesterol, d-α-tocopheryl polyethylene glycol 1000
succinate-triphenylphosphine conjugate and egg phosphatidylcholine in the molar
ratio of 3.5:8.5:88 respectively. In lung cancer, the above-formulated paclitaxel
liposomes showed apoptosis in drug-resistant cancer cells via stimulation of pro-
apoptotic proteins such as Bid and Bax and suppression of anti-apoptotic protein
Bcl-2 [34]. Daunorubicin (Fig. 1), which is extracted from Streptomyces peucetius,
works by concentrating inside the mammary adenocarcinoma cells and murine lym-
phosarcoma cells. The uptake of this agent gets enhanced between 2.5 and 20 times
when formulated as a liposomal drug delivery system, in comparison to free dauno-
rubicin [37, 38]. Different types of liposomes that have been studied to carry cyto-
toxic phytochemicals are discussed in Table 2.
3 Carbon Nanotubes
Carbon nanotubes (CNTs), also known as tubular fullerenes [44], were first discov-
ered in 1991 by Japanese scientist Iijima [45]. CNTs are arising as novel nanomate-
rials for different biomedical applications and successfully developed to deliver
different medicinal agents and biomolecules to targeted sites. The lower cytotoxic-
ity and biocompatibility depend on dose, testing system, size, duration and surface
functionalization. The potential drawback of these systems is high cytotoxicity, lim-
iting their applications in different biological systems and humans. However, the
solubility and biocompatibility improvement for reducing cytotoxicity may be per-
formed by functionalization of CNTs [46]. CNTs and their connected materials
have numerous trademark properties and applications, settling on them an
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1371
appropriate decision in the biomedical field, for example, drug release, gene ther-
apy, drug release and tissue engineering [47]. CNTs may have a length-to-diameter
ratio greater than 1,000,000 [48]. In CNTs, the medicinal components can be
entrapped in rolled graphene (with SP2 hybridization) sheets because of which it
gains more extensive length, high surface area and narrow diameter. The surface
modifications, e.g. attaching oxygen containing groups over the surface signifi-
cantly contribution in ligand targeting [49]. The ‘single-walled carbon nanotubes
(SWCNTs)’ and ‘multi-wall carbon nanotubes (MWCNTs)’ are contemplated to be
folded graphene sheets with lengths going from ~50 to 1000 nm and widths of ~5 to
20 nm [50]. Recently, attempts were made by researchers to extent the applicability
of CNTs for diagnosis, drug targeting and viral protein detection for fighting against
for different types of viral infections including corona virus and HIV [51]. Various
properties result from the development of carbon molecules in graphene chambers.
CNTs are tremendous round and hollow huge particle comprising of a hexagonal
arrangement of sp2 hybridized carbon atoms (C-C distance 1.4 Ǻ) [52]. The
researchers have found significant increase in efficiency in the perspective of tar-
geted tumour accumulation by functionalized SWNTs with targeting ligands RGD
peptide or antibodies [53]. The research published by Li et al. [54] describes
SWCNT system of curcumin, a polyphenolic anti-cancerous compound with poor
bioavailability. Polyvinylpyrrolidone and phosphatidylcholine were used for sur-
face modification increasing the concentration (18×) of curcumin in blood in mice.
The coating also increases the stability of curcumin and improving the shortfall of
drug resistance. The overall result indicates increase in uptake of curcumin in
human prostate cancerous PC-3 cells with promising inhibition of tumour growth.
The nSiO2 microspheres after inhalation can cross blood–lung barrier and enters
into blood circulation possibly producing similar to systemic administration. These
can also be administered through injections or percutaneous permeation [67]. At the
point when magnetic particles are encircled by phospholipid bilayers, a colloidal
construction is shaped, which is generally depicted as ‘magnetic liposomes’. The
interior pit of this kind of liposome is totally involved by iron oxide particles [68].
MNPs are inclined to agglomeration because of high surface energy and huge
explicit surface area, making it hard to consistently scatter in polymers, which
incredibly restricts their exploration and application. Along these lines, polymer
change is a viable method for working on this peculiarity [69]. In the study of Tousi
et al. [70], the anticancerous activity of Eupatorin (Fig. 1), a natural methoxyfla-
vone, when encapsulated in ‘Fe3O4@mPEG-b-PLGA’ nanoparticles, was found to
be increased in prostate cancer cell lines as compared to free molecule. The research-
ers used oleic acid for stabilization of Fe3O4 nanoparticles were with the ‘mPEG-b-
PLGA’ polymer for increasing cell uptake and its biocompatibility. The ‘poly
(lactic-co-glycolic) acid (PLGA)’ was used because of its biocompatibility and
releasing nontoxic monomers and oligomers, glycolic and lactic acid. The study
also found the formulation decreasing the necrosis and improved apoptosis which
may be an alternative for treating drug resistant cancer. In another publication, inhi-
bition activity against MCF-7 breast cancer cells and KB nasopharyngeal cancer
cells was observed using folate-grafted curcumin-loaded magnetic nanoparticles
reported by Montazerabadi et al. [71]. Upon alteration of magnetic field, the MNPs
can generate localized heat which are used to mediate cancer hyperthermia [72].
Montazerabadi et al. claim that problems associated with curcumin like low water
solubility, very poor bioavailability, rapid systemic elimination and metabolism
may be overcome by using their developed nanoparticles [71].
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1373
5 Micelles
These are amphiphilic surfactant drug delivery system which shows the property of
aggregation by forming spherical vesicles when getting in contact with water. The
hydrophobic drugs can be incorporated into the inner nucleus of micelle which is
also hydrophobic. The basic structure of a micelle is a hydrophobic tail and a hydro-
philic head. The tail is composed of long chain fatty acids and the molecular weight
is greater as compared to other nanoparticle delivery system which makes it an ideal
choice for tissue targeting in case of solid tumours [73]. In a study conducted by Qiu
et al. synthesized polymeric micelles carrying luteolin (Fig. 1) and tested them on
C-26 colon cancer cell line. In rats the comparison was drawn between the formu-
lated preparation and free luteolin, it was noted that the luteolin polymeric micelles
lead to enhanced cytotoxic and bioavailability of the drug [74]. In another synthesis
study carried out by Dong et al. encapsulated honokiol (Fig. 1) in a star-shaped
polymeric micelle composed of poly(3-caprolactone) and monomethoxy
poly(ethylene glycol) via dissolution ultrasonic technique and tested the cytotoxic
profile on murine colon carcinoma CT26 cell lines. It was concluded that the poly-
meric micelles showed dose-related cytotoxic on the cancer cell lines [75]. The
honokiol micelles were even synthesized by Wei et al. which were composed of
copolymer poly(3-caprolactone)-poly(ethylene glycol)-poly(3-caprolactone) and
was tested in human lung adenocarcinoma cell line A549. The antiproliferative
action of the micelles were enhanced as compared to the free drug when tested in
the A549 cell lines [76]. Guo X formulated polymeric micelles composed of resve-
ratrol (Fig. 1) and semi-synthetic drug docetaxel (Fig. 1) for the treatment of drug-
resistant breast cancer. For their formulation, they used methoxyl poly(ethylene
glycol)-poly(d,l-lactide) copolymer and were tested against MCF-7 breast cancer
cell lines. Prolonged drug release was observed with increased cytotoxicity as com-
pared to free drug [77].
6 Dendrimers
increased cytotoxic profile on the breast cancer cell lines [80]. Various types of den-
drimers that are used to carry plant-based anticancer drug are discussed in Table 4.
These are colloidal drug carriers and were developed as an alternative to previous
drug delivery agents such as polymeric nanoparticles, liposomes and emulsions.
These are next generation of submicron-sized lipid emulsion in which the oil, called
as liquid lipid, has been replaced by solid lipid. These nanocarriers have increased
surface area, enhanced drug loading capacity and are smaller in size [87]. These are
made from using the biodegradable polymers that have low acidity and toxicity, as
majority of the cytotoxic compounds are lipophilic and thus, they can be incorpo-
rated into this drug delivery system, making oral administration possible for cyto-
toxic drug [88]. In a study conducted by Serpe et al. formulated paclitaxel solid lipid
nanoparticles which showed similar cytotoxic profile as compared to drug present
in free solution. The efficacy of the developed formulation was tested on breast
cancer murine model and it was noted that the animals which received solid lipid
nanoparticles containing paclitaxel had reduced tumour size [89]. Solid lipid
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1375
8 Exosomes
These are extracellular vesicles with a diameter of 30–150 nm and are secreted by B
cells, macrophages, T cells, dendritic cells, endothelial cells, epithelial cells and
endothelial cells. Exosomes are employed as a drug delivery agent in cancer research
because they have low immunogenicity, biocompatibility and biodistribution [91].
Different studies have highlighted that paclitaxel exosomes can amplify the clinical
effectiveness of exosomes. A study was conducted in which exosomes were extracted
from milk and were equipped with paclitaxel for testing them against xenografts of
human lung tumour. The prepared novel drug delivery system showed less immune
system toxicity in comparison to free drug [92]. Exosomes extracted from U-87 cell
line have the capability of passing the blood–brain barrier and they can be equipped
with paclitaxel to target glioblastoma multiforme [93]. It was noted from various
studies that the stability and bioavailability curcumin was by using exosomes as its
carrier. These exosomes were tested in GL26 brain tumour model and were given
via the intranasal route. The effect was reduction in inflammation, delayed in tumour
growth and mitigation of the brain endothelial cells [94]. In case of pancreatic can-
cer cells, the curcumin-exosome complex reduced the proliferation of cancer and
caused induction of apoptosis [95, 96]. Celastrol (Fig. 1), a plant-based triterpenoid,
has shown apoptosis activity against multidrug-resistant oral cancer cells from vin-
cristine (Fig. 1) via the JNK1/2 signalling pathway [97]. It was complexed with the
milk-derived exosomes and were tested in vitro against lung cancer. These were
proved to be stable, having oral delivery and low systemic toxicity [98]. Certain
agents such as docosahexaenoic acid (Fig. 1) can enhance the secretion of exosomes
and raise the small RNA levels for inhibition of pro-angiogenesis mRNAs which
lead to suppression of cancer angiogenesis in case of breast cancer [99].
9 Polymeric Nanoparticles
These range in the size between 10 nm and 1000 nm and have beneficial properties
such as enhanced therapeutic effect of the encapsulated drug, site specificity and
route of administration. These are mostly used in ophthalmic and oral administration
due to increased bioavailability and reduced side effects [100]. In cytotoxic drug
delivery, polymeric nanoparticles provide a barrier between the anti-cancer drug and
1376 T. Alex et al.
the healthy cells, thus reducing the toxicity profile of the carrier drug [101]. Bisht
et al. prepared polymeric nanoparticles carrying curcumin and they were composed
of crosslinked copolymers of N-vinyl-2-pyrrolidone, poly(ethylene glycol) monoac-
rylate and N-isopropylacrylamide. This nanodrug delivery system showed effective-
ness against human pancreatic tumour cell line and it induced apoptosis, regulated
levels of proinflammatory cytokines and blocks the activation of nuclear factor
kappa B [102]. Various plant-based cytotoxic drugs that have been formulated as a
polymeric nanoparticle drug delivery system are discussed in Table 5.
10 Nanofibres
This novel drug delivery system is synthesized using electrospinning technique, and
the formulated nanofibres are biodegradable as well as biocompatible. Nanofibres
have high surface area to mass ratio and low density [110]. Exceptional anti-cancer
activity was achieved in breast cancer model where camptothecin (Fig. 1) was com-
plexed with peptide amphiphile nanofibres [111]. To establish the efficacy of camp-
tothecin in lung cancer, nanofibres loaded with DNA were used to carry the molecule
and showed effective drug loading effect [112]. Vashisth et al. developed
poly(lactide-co-glycolide)–polycaprolactone biodegradable nanofibres carrying
quercetin (Fig. 1) and tested them for their efficacy on human hepatocellular cancer
cells. The novel preparation showed sustained release of quercetin along with
enhanced diffusion and aqueous permeability. The drug-releasing pattern initially
was a blast released, followed by slow release kinetics [113]. In another study con-
ducted by Gajanan et al., incorporated Emblica officinalis into the nanofibres made
up of polycaprolactone. The prepared formulation showed antiproliferative action
on human breast cancer cell lines [114]. Different types of nanofibres that are used
for carrying plant-based cytotoxic drugs are discussed in Table 6.
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1377
In past two decades, it has been well established that nanocarriers are equipped with
advantages such as protecting the cytotoxic phytochemical from potential degrada-
tion, enhanced pharmacokinetic profile of the active drug and greater drug absorp-
tion at tumour site. The accumulation of nanoparticles at the tumour site is due to
the enhanced permeability effect [121]. The various nanocarriers that have been
approved by FDA in past are discussed in Table 7 and nanocarriers that are undergo-
ing clinical trials are discussed in Table 8 [122].
Table 8 Clinical trial status of various nanocarriers carrying cytotoxic phytochemicals
1378
12 Conclusion
Systemic toxicities and drug resistance are the major problems associated with the
synthetic and semi-synthetic anticancer drugs. Nowadays, due to new extraction
and separation techniques and availability of high sophisticated machines, various
potent anticancer molecules are being extracted from plants. However, these phyto-
chemicals have short half-life, nonselective biodistribution, poor water solubility,
most of the agents are hydrophobic with low bioavailability which leads to hin-
drance in their clinical application. To subdue these effects, various nanocarriers
such as liposomes, dendrimers, micelles, magnetic nanoparticles, carbon nanotubes,
exosomes, polymeric nanoparticles, nanofibre and solid lipid nanoparticles can be
used to encapsulate the plant-based cytotoxic agents. The systemic toxicity is
reduced along with enhanced pharmacological activity and bioavailability.
References
1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A (2018) Global cancer sta-
tistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in
185 countries. CA Cancer J Clin 68(6):394–424
2. Nussbaumer S, Bonnabry P, Veuthey JL, Fleury-Souverain S (2011) Analysis of anticancer
drugs: a review. Talanta 85(5):2265–2289
3. Amin A, Gali-Muhtasib H, Ocker M, Schneider-Stock R (2009) Overview of major classes of
plant-derived anticancer drugs. Int J Biomed Sci 5(1):1
4. Che E, Gao Y, Wan L, Zhang Y, Han N, Bai J, Li J, Sha Z, Wang S (2015) Paclitaxel/gelatin
coated magnetic mesoporous silica nanoparticles: preparation and antitumor efficacy in vivo.
Microporous Mesoporous Mater 204:226–234
5. Singh A, Menéndez-Perdomo IM, Facchini PJ (2019) Benzylisoquinoline alkaloid biosyn-
thesis in opium poppy: an update. Phytochem Rev 18(6):1457–1482
6. Shah U, Shah R, Acharya S, Acharya N (2013) Novel anticancer agents from plant sources.
Chin J Nat Med 11(1):16–23
7. Cragg GM, Newman DJ (2005) Plants as a source of anti-cancer agents. J Ethnopharmacol
100(1–2):72–79
8. Son IH, Chung IM, Lee SI, Yang HD, Moon HI (2007) Pomiferin, histone deacetylase inhibitor
isolated from the fruits of Maclura pomifera. Bioorganic Med Chem Lett 17(17):4753–4755
9. Cornblatt BS, Ye L, Dinkova-Kostova AT, Erb M, Fahey JW, Singh NK, Chen MS, Stierer T,
Garrett-Mayer E, Argani P, Davidson NE (2007) Preclinical and clinical evaluation of sul-
foraphane for chemoprevention in the breast. Carcinogenesis 28(7):1485–1490
10. Hadjzadeh M, Tavakol Afshari J, Ghorbani A, Shakeri MT (2006) The effects of aque-
ous extract of garlic (Allium sativum L.) on laryngeal cancer cells (Hep-2) and L929 cells
in vitro. J Med Plants 5(18):41–48
11. Sadeghnia HR, Ghorbani Hesari T, Mortazavian SM, Mousavi SH, Tayarani-Najaran Z,
Ghorbani A (2014) Viola tricolor induces apoptosis in cancer cells and exhibits antiangio-
genic activity on chicken chorioallantoic membrane. Biomed Res Int 2014:625792
12. Sharifi-Rad J, Quispe C, Patra JK, Singh YD, Panda MK, Das G, Adetunji CO, Michael
OS, Sytar O, Polito L, Živković J (2021) Paclitaxel: application in modern oncology and
nanomedicine-based cancer therapy. Oxi Med Cell Longev 2021:3687700
1380 T. Alex et al.
13. Mousa SA, Bharali DJ (2011) Nanotechnology-based detection and targeted therapy in can-
cer: nano-bio paradigms and applications. Cancers 3(3):2888–2903
14. Nie S, Xing Y, Kim GJ, Simons JW (2007) Nanotechnology applications in cancer. Annu Rev
Biomed Eng 9:257–288
15. Cho K, Wang XU, Nie S, Shin DM (2008) Therapeutic nanoparticles for drug delivery in
cancer. Clin Cancer Res 14(5):1310–1316
16. Nikitin MP, Zelepukin IV, Shipunova VO, Sokolov IL, Deyev SM, Nikitin PI (2020)
Enhancement of the blood-circulation time and performance of nanomedicines via the forced
clearance of erythrocytes. Nat Biomed Eng 4(7):717–731
17. Ahmed A, Sarwar S, Hu Y, Munir MU, Nisar MF, Ikram F, Asif A, Rahman SU, Chaudhry
AA, Rehman IU (2021) Surface-modified polymeric nanoparticles for drug delivery to cancer
cells. Expert Opin Drug Deliv 18(1):1–24
18. Jain RK, Stylianopoulos T (2010) Delivering nanomedicine to solid tumors. Nat Rev Clin
Oncol 7(11):653–664
19. Ding Y, Xu Y, Yang W, Niu P, Li X, Chen Y, Li Z, Liu Y, An Y, Liu Y, Shen W (2020)
Investigating the EPR effect of nanomedicines in human renal tumors via ex vivo perfusion
strategy. Nano Today 35:100970
20. Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, Wu S, Deng Y, Zhang J, Shao A (2020)
Nanoparticle-based drug delivery in cancer therapy and its role in overcoming drug resis-
tance. Front Mol Biosci 7:193
21. Ingale AG, Chaudhari AN (2013) Biogenic synthesis of nanoparticles and potential applica-
tions: an eco-friendly approach. J Nanomed Nanotechol 4(165):1–7
22. Chauhan RP, Gupta C, Prakash D (2012) Methodological advancements in green nano-
technology and their applications in biological synthesis of herbal nanoparticles. Int J
Bioassays 01:6–10
23. Parveen K, Banse V, Ledwani L (2016) Green synthesis of nanoparticles: their advantages
and disadvantages. In: AIP conference proceedings, vol 1724(1). AIP Publishing LLC,
Melville, New York, United States. pp 020048
24. Kuang Y, Wang Q, Chen Z, Megharaj M, Naidu R (2013) Heterogeneous Fenton-like oxida-
tion of monochlorobenzene using green synthesis of iron nanoparticles. J Colloid Interface
Sci 410:67–73
25. Tripathi N, Pavelyev V, Islam SS (2017) Synthesis of carbon nanotubes using green plant
extract as catalyst: unconventional concept and its realization. Appl Nanosci 7(8):557–566
26. Alippilakkotte S, Kumar S, Sreejith L (2017) Fabrication of PLA/Ag nanofibers by green
synthesis method using Momordica charantia fruit extract for wound dressing applications.
Colloids Surf A Physicochem Eng Asp 529:771–782
27. Frolov VA, Shnyrova AV, Zimmerberg J (2011) Lipid polymorphisms and membrane shape.
Cold Spring Harb Perspect Biol 3(11):a004747
28. Drummond DC, Meyer O, Hong K, Kirpotin DB, Papahadjopoulos D (1999) Optimizing
liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacol Rev
51(4):691–744
29. Matsumura Y, Maeda H (1986) A new concept for macromolecular therapeutics in cancer
chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent
smancs. Cancer Res 46(12 Part 1):6387–6392
30. Oussoren C, Eling WM, Crommelin DJ, Storm G, Zuidema J (1998) The influence of the
route of administration and liposome composition on the potential of liposomes to protect tis-
sue against local toxicity of two antitumor drugs. Biochim Biophys Acta (BBA)-Biomembr
1369(1):159–172
31. Sawant RR, Torchilin VP (2010) Liposomes as ‘smart’pharmaceutical nanocarriers. Soft
Matter 6(17):4026–4044
32. Feng T, Wei Y, Lee RJ, Zhao L (2017) Liposomal curcumin and its application in cancer. Int
J Nanomedicine 12:6027
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1381
33. Li PM, Li YL, Liu B, Wang WJ, Wang YZ, Li Z (2014) Curcumin inhibits MHCC97H liver
cancer cells by activating ROS/TLR-4/caspase signaling pathway. Asian Pac J Cancer Prev
15(5):2329–2334
34. Zhou J, Zhao WY, Ma X, Ju RJ, Li XY, Li N, Sun MG, Shi JF, Zhang CX, Lu WL (2013) The
anticancer efficacy of paclitaxel liposomes modified with mitochondrial targeting conjugate
in resistant lung cancer. Biomaterials 34(14):3626–3638
35. Xu X, Wang L, Xu HQ, Huang XE, Qian YD, Xiang J (2013) Clinical comparison between
paclitaxel liposome (Lipusu®) and paclitaxel for treatment of patients with metastatic gastric
cancer. Asian Pac J Cancer Prev 14(4):2591–2594
36. Surapaneni MS, Das SK, Das NG (2012) Designing Paclitaxel drug delivery systems aimed
at improved patient outcomes: current status and challenges. Int Sch Res Notices 2012:1–15
37. Forssen EA, Coulter DM, Proffitt RT (1992) Selective in vivo localization of daunorubicin
small unilamellar vesicles in solid tumors. Cancer Res 52(12):3255–3261
38. Klein K, Kaspers GL (2013) A review of liposomal daunorubicin in the treatment of acute
leukemia. Oncol Hematol Rev 9(2):142–148
39. Feldman EJ, Lancet JE, Kolitz JE, Ritchie EK, Roboz GJ, List AF, Allen SL, Asatiani E,
Mayer LD, Swenson C, Louie AC (2011) First-in-man study of CPX-351: a liposomal car-
rier containing cytarabine and daunorubicin in a fixed 5: 1 molar ratio for the treatment of
relapsed and refractory acute myeloid leukemia. J Clin Oncol 29(8):979
40. O’Byrne KJ, Thomas AL, Sharma RA, DeCatris M, Shields F, Beare S, Steward WP (2002)
A phase I dose-escalating study of DaunoXome, liposomal daunorubicin, in metastatic breast
cancer. Br J Cancer 87(1):15–20
41. Li XT, Ju RJ, Li XY, Zeng F, Shi JF, Liu L, Zhang CX, Sun MG, Lou JN, Lu WL (2014)
Multifunctional targeting daunorubicin plus quinacrine liposomes, modified by wheat germ
agglutinin and tamoxifen, for treating brain glioma and glioma stem cells. Oncotarget
5(15):6497
42. Du R, Zhong T, Zhang WQ, Song P, Song WD, Zhao Y, Wang C, Tang YQ, Zhang X, Zhang
Q (2014) Antitumor effect of iRGD-modified liposomes containing conjugated linoleic acid–
paclitaxel (CLA-PTX) on B16-F10 melanoma. Int J Nanomedicine 9:3091
43. Hu L, Liang G, Yuliang W, Bingjing Z, Xiangdong Z, Rufu X (2013) Assessing the effective-
ness and safety of liposomal paclitaxel in combination with cisplatin as first-line chemother-
apy for patients with advanced NSCLC with regional lymph-node metastasis: study protocol
for a randomized controlled trial (PLC-GC trial). Trials 14(1):1–7
44. Ibrahim KS (2013) Carbon nanotubes-properties and applications: a review. Carbon Lett
14(3):131–144
45. Patel J, Parikh S, Patel S, Patel R, Patel P (2021) Carbon nanotube (CNTs): structure, syn-
thesis, purification, functionalisation, pharmacology, toxicology, biodegradation and appli-
cation as nanomedicine and biosensor: carbon nanotube (CNTs). J Pharm Sci Med Res
1(02):017–044
46. Vardharajula S, Ali SZ, Tiwari PM, Eroğlu E, Vig K, Dennis VA, Singh SR (2012)
Functionalized carbon nanotubes: biomedical applications. Int J Nanomedicine 7:5361–5374
47. Thakur A, Bharti R, Sharma R (2021) Carbon nanotubes: types, synthesis, cytotoxicity and
applications in biomedical. Mater Today Proceed 50:2256–2268
48. Rahamathulla M, Bhosale RR, Osmani RAM, Mahima KC, Johnson AP, Hani U, Ghazwani
M, Begum MY, Alshehri S, Ghoneim MM, Shakeel F, Gangadharappa HV (2021) Carbon
nanotubes: current perspectives on diverse applications in targeted drug delivery and thera-
pies. Materials (Basel) 14(21):6707
49. Son KH, Hong JH, Lee JW (2016) Carbon nanotubes as cancer therapeutic carriers and medi-
ators. Int J Nanomedicine 11:5163–5185
50. Rashid MH, Ralph SF (2017) Carbon nanotube membranes: synthesis, properties, and future
filtration applications. Nanomaterials (Basel) 7(5):99
51. Varghese R, Salvi S, Sood P, Karsiya J, Kumar D (2022) Carbon nanotubes in COVID-19: a
critical review and prospects. Colloid Interface Sci Commun 46:100544
1382 T. Alex et al.
52. Kushwaha SK, Ghoshal S, Rai AK, Singh S (2013) Carbon nanotubes as a novel drug deliv-
ery system for anticancer therapy: a review. Braz J Pharm Sci 49(4):629–643
53. Liu Z, Chen K, Davis C, Sherlock S, Cao Q, Chen X, Dai H (2008) Drug delivery with carbon
nanotubes for in vivo cancer treatment. Cancer Res 68(16):6652–6660
54. Li H, Zhang N, Hao Y, Wang Y, Jia S, Zhang H (2019) Enhancement of curcumin antitumor
efficacy and further photothermal ablation of tumor growth by single-walled carbon nano-
tubes delivery system in vivo. Drug Deliv 26(1):1017–1026
55. Ali A, Shah T, Ullah R, Zhou P, Guo M, Ovais M, Tan Z, Rui Y (2021) Review on recent
progress in magnetic nanoparticles: synthesis, characterization, and diverse applications.
Front Chem 9:629054
56. Liu Y, Guo Z, Li F, Xiao Y, Zhang Y, Bu T, Jia P, Zhe T, Wang L (2019) Multifunctional
magnetic copper ferrite nanoparticles as fenton-like reaction and near-infrared photothermal
agents for synergetic antibacterial therapy. ACS Appl Mater Interfaces 11(35):31649–31660
57. Chmykhalo V, Belanova A, Belousova M, Butova V, Makarenko Y, Khrenkova V, Soldatov
A, Zolotukhin P (2021) Microbial-based magnetic nanoparticles production: a mini-review.
Integr Biol (Camb) 13(4):98–107
58. Malar GCG, Seenuvasan M, Kumar KS, Kumar MA (2021) Instrumental methods in sur-
face property analysis of magnetic nanoparticles. In: Kumar RP, Bharathiraja B (eds)
Nanomaterials. Academic Press, Cambridge, Massachusetts, United States, pp 691–697
59. Alonso J, Barandiarán JM, Fernández Barquín L, García-Arribas A (2018) Magnetic nanopar-
ticles, synthesis, properties, and applications. In: El-Gendy AA, Barandiarán JM, Hadimani
RL (eds) Micro and nano technologies, magnetic nanostructured materials. Elsevier, pp 1–40
60. Li X, Li W, Wang M, Liao Z (2021) Magnetic nanoparticles for cancer theranostics: advances
and prospects. J Control Release 335:437–448
61. Iler KR (1979) The chemistry of silica. Solubility, polymerization, colloid and surface prop-
erties and biochemistry of silica. Wiley, Chichester
62. Do Kim K, Kim SS, Choa YH, Kim HT (2007) Formation and surface modification of Fe3O4
nanoparticles by co-precipitation and sol-gel method. J Ind Eng Chem 13(7):1137–1141
63. Jafarzadeh M, Soleimani E, Sepahvand H, Adnan R (2015) Synthesis and characterization
of fluconazole-functionalized magnetic nanoparticles as a catalyst for the synthesis of 3-aryl
and 3-amino-imidazo [1, 2-a] pyridines. RSC Adv 5(53):42744–42753
64. Oliveira RR, Carrião MS, Pacheco MT, Branquinho LC, de Souza ALR, Bakuzis AF, Lima
EM (2018) Triggered release of paclitaxel from magnetic solid lipid nanoparticles by mag-
netic hyperthermia. Mater Sci Eng C Mater Biol Appl 92:547–553
65. Altenschmidt L, Sánchez-Paradinas S, Lübkemann F, Zámbó D, Abdelmonem AM,
Bradtmüller H, Masood A, Morales I, de la Presa P, Knebel A, García-Tuñón MAG, Pelaz
B, Hindricks KDJ, Behrens P, Parak WJ, Bigall NC (2021) Aerogelation of polymer-coated
photoluminescent, plasmonic, and magnetic nanoparticles for biosensing applications. ACS
Appl Nano Mater 4(7):6678–6688
66. Talluri S, Malla RR (2019) Superparamagnetic iron oxide nanoparticles (SPIONs) for diag-
nosis and treatment of breast, ovarian and cervical cancers. Curr Drug Metab 20(12):942–945
67. Hou H, Wang C, Nan K, Freeman WR, Sailor MJ, Cheng L (2016) Controlled release of
dexamethasone from an intravitreal delivery system using porous silicon dioxide. Invest
Ophthalmol Vis Sci 57(2):557–566. https://doi.org/10.1167/iovs.15-18559
68. Diksha RI (2012) Synthesis, surface modification, characterization, and biomedical in vitro
applications of organically modified silica (ORMOSIL) nanoparticles. Methods Mol Biol
906:365–379
69. Zhang M, Qiao J, Qi L (2018) Dual-functional polymer-modified magnetic nanoparticles for
isolation of lysozyme. Anal Chim Acta 1035:70–76
70. Tousi MS, Sepehri H, Khoee S, Farimani MM, Delphi L, Mansourizadeh F (2021) Evaluation
of apoptotic effects of mPEG-b-PLGA coated iron oxide nanoparticles as a eupatorin carrier
on DU-145 and LNCaP human prostate cancer cell lines. J Pharm Anal 11(1):108–121
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1383
91. Bang C, Thum T (2012) Exosomes: new players in cell–cell communication. Int J Biochem
Cell Biol 44(11):2060–2064
92. Agrawal AK, Aqil F, Jeyabalan J, Spencer WA, Beck J, Gachuki BW, Alhakeem SS, Oben
K, Munagala R, Bondada S, Gupta RC (2017) Milk-derived exosomes for oral delivery of
paclitaxel. Nanomedicine 13(5):1627–1636
93. Salarpour S, Forootanfar H, Pournamdari M, Ahmadi-Zeidabadi M, Esmaeeli M, Pardakhty
A (2019) Paclitaxel incorporated exosomes derived from glioblastoma cells: comparative
study of two loading techniques. DARU J Pharm Sci 27(2):533–539
94. Zhuang X, Xiang X, Grizzle W, Sun D, Zhang S, Axtell RC, Ju S, Mu J, Zhang L, Steinman
L, Miller D (2011) Treatment of brain inflammatory diseases by delivering exosome encapsu-
lated anti-inflammatory drugs from the nasal region to the brain. Mol Ther 19(10):1769–1779
95. Osterman CJ, Lynch JC, Leaf P, Gonda A, Ferguson Bennit HR, Griffiths D, Wall NR (2015)
Curcumin modulates pancreatic adenocarcinoma cell-derived exosomal function. PLoS One
10(7):e0132845
96. Wei H, Chen J, Wang S, Fu F, Zhu X, Wu C, Liu Z, Zhong G, Lin J (2019) A nanodrug con-
sisting of doxorubicin and exosome derived from mesenchymal stem cells for osteosarcoma
treatment in vitro. Int J Nanomedicine 14:8603
97. Lin FZ, Wang SC, Hsi YT, Lo YS, Lin CC, Chuang YC, Lin SH, Hsieh MJ, Chen MK (2019)
Celastrol induces vincristine multidrug resistance oral cancer cell apoptosis by targeting
JNK1/2 signaling pathway. Phytomedicine 54:1–8
98. Aqil F, Kausar H, Agrawal AK, Jeyabalan J, Kyakulaga AH, Munagala R, Gupta R (2016)
Exosomal formulation enhances therapeutic response of celastrol against lung cancer. Exp
Mol Pathol 101(1):12–21
99. Hannafon BN, Carpenter KJ, Berry WL, Janknecht R, Dooley WC, Ding WQ (2015)
Exosome-mediated microRNA signaling from breast cancer cells is altered by the anti-
angiogenesis agent docosahexaenoic acid (DHA). Mol Cancer 14(1):1–3
100. Schaffazick SR, Guterres SS, Freitas LD, Pohlmann AR (2003) Caracterização e estabilidade
físico-química de sistemas poliméricos nanoparticulados para administração de fármacos.
Química nova 26:726–737
101. Brewer E, Coleman J, Lowman A (2011) Emerging technologies of polymeric nanoparticles
in cancer drug delivery. J Nanomater 2011:1–10
102. Bisht S, Feldmann G, Soni S, Ravi R, Karikar C, Maitra A, Maitra A (2007) Polymeric
nanoparticle-encapsulated curcumin (“nanocurcumin”): a novel strategy for human cancer
therapy. J Nanobiotech 5(1):1–8
103. Khanna V, Kalscheuer S, Kirtane A, Zhang W, Panyam J (2019) Perlecan-targeted nanopar-
ticles for drug delivery to triple-negative breast cancer. Future Drug Discov 1(1):FDD8
104. Shafiei-Irannejad V, Samadi N, Salehi R, Yousefi B, Rahimi M, Akbarzadeh A, Zarghami N
(2018) Reversion of multidrug resistance by co-encapsulation of doxorubicin and metformin
in poly (lactide-co-glycolide)-d-α-tocopheryl polyethylene glycol 1000 succinate nanopar-
ticles. Pharma Res 35(6):1–3
105. Yuan JD, ZhuGe DL, Tong MQ, Lin MT, Xu XF, Tang X, Zhao YZ, Xu HL (2018) pH-
sensitive polymeric nanoparticles of mPEG-PLGA-PGlu with hybrid core for simultaneous
encapsulation of curcumin and doxorubicin to kill the heterogeneous tumour cells in breast
cancer. Artif Cells Nanomed Biotechnol 46(sup1):302–313
106. Jin M, Jin G, Kang L, Chen L, Gao Z, Huang W (2018) Smart polymeric nanoparticles
with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin
siRNA to enhance antitumor outcomes. Int J Nano 13:2405
107. Çırpanlı Y, Allard E, Passirani C, Bilensoy E, Lemaire L, Çalış S, Benoit JP (2011)
Antitumoral activity of camptothecin-loaded nanoparticles in 9L rat glioma model. Int J
Pharma 403(1–2):201–206
108. Soma CE, Dubernet C, Bentolila D, Benita S, Couvreur P (2000) Reversion of multidrug
resistance by co-encapsulation of doxorubicin and cyclosporin A in polyalkylcyanoacrylate
nanoparticles. Biomaterials 21(1):1–7
Nanotechnological Modus Operandi for the Delivery of Cytotoxic Phytochemicals 1385
109. Zhao Y, Cai C, Liu M, Zhao Y, Pei W, Chu X, Zhang H, Wang Z, Han J (2019) An organic
solvent-free technology for the fabrication of albumin-based paclitaxel nanoparticles for
effective cancer therapy. Colloids Surf B: Biointerfaces 183:110394
110. Yu Y, Kong L, Li L, Li N, Yan P (2015) Antitumor activity of doxorubicin-loaded carbon
nanotubes incorporated poly (lactic-co-glycolic acid) electrospun composite nanofibers.
Nanoscale Res Lett 10(1):1–9
111. Soukasene S, Toft DJ, Moyer TJ, Lu H, Lee HK, Standley SM, Cryns VL, Stupp SI (2011)
Antitumor activity of peptide amphiphile nanofiber-encapsulated camptothecin. ACS Nano
5(11):9113–9121
112. Baig MM, Lai WF, Ashraf S, Saleem A, Akhtar MF, Mikrani R, Naveed M, Siddique F,
Taleb A, Mudassir J, Khan GJ (2020) The integrin facilitated internalization of fibronectin-
functionalized camptothecin-loaded DNA-nanofibers for high-efficiency anticancer effects.
Drug Deliv Transl Res 10(5):1381–1392
113. Vashisth P, Singh RP, Pruthi V (2016) A controlled release system for quercetin from biode-
gradable poly (lactide-co-glycolide)–polycaprolactone nanofibers and its in vitro antitumor
activity. J Bioact Compat Polym 31(3):260–272
114. Arbade GK, Kumar V, Tripathi V, Menon A, Bose S, Patro TU (2019) Emblica officinalis-
loaded poly (ε-caprolactone) electrospun nanofiber scaffold as potential antibacterial and
anticancer deployable patch. New J Chem 43(19):7427–7440
115. Amna T, Barakat NA, Hassan MS, Khil MS, Kim HY (2013) Camptothecin loaded poly
(ε-caprolactone) nanofibers via one-step electrospinning and their cytotoxicity impact.
Colloids Surf A Physicochem Eng Asp 431:1–8
116. Zhou Z, Piao Y, Hao L, Wang G, Zhou Z, Shen Y (2019) Acidity-responsive shell-
sheddable camptothecin-based nanofibers for carrier-free cancer drug delivery. Nanoscale
11(34):15907–15916
117. Sedghi R, Shaabani A, Mohammadi Z, Samadi FY, Isaei E (2017) Biocompatible electro-
spinning chitosan nanofibers: a novel delivery system with superior local cancer therapy.
Carbohydr Polym 159:1
118. Wen P, Hu TG, Li L, Zong MH, Wu H (2018) A colon-specific delivery system for quer-
cetin with enhanced cancer prevention based on co-axial electrospinning. Food Funct
9(11):5999–6009
119. Eskitoros-Togay ŞM, Bulbul YE, Dilsiz N (2018) Quercetin-loaded and unloaded electro-
spun membranes: synthesis, characterization and in vitro release study. J Drug Deliv Sci
Technol 47:22–30
120. Stoyanova N, Spasova M, Manolova N, Rashkov I, Georgieva A, Toshkova R (2020)
Antioxidant and antitumor activities of novel quercetin-loaded electrospun cellulose acetate/
polyethylene glycol fibrous materials. Antioxidants 9(3):232
121. Vieira DB, Gamarra LF (2016) Advances in the use of nanocarriers for cancer diagnosis and
treatment. Einstein (Sao Paulo) 14:99–103
122. ClinicalTrials.gov. https://clinicaltrials.gov/. Accessed 03 Feb 2022
Plant-Based Green Nanoparticles
in Cancer Diagnosis and Chemotherapy
1 Introduction
A. John (*)
St. Thomas College, Kozhencherry, Kerala, India
R. E. Roy
Christian College, Alappuzha, Kerala, India
biochemical and optical properties of some nanomaterials help in rapid and sensi-
tive detection of cancer cells and tumours [2]. Nanoparticles act as selective drug
delivery vehicles, deliver medicines like chemotherapy directly to the tumours.
Even though the nanoparticles are in the 1–100 nm size range these can also be
classified on the basis of the nanostructure dimensions as zero-dimensional (0D),
one-dimensional (1D), two-dimensional (2D) and three-dimensional (3D) nanoma-
terials. Zero-dimensional nanomaterials are nanoparticles that have nanodimension
in all the three dimensions, for example, quantum dots, metal nanoparticles, carbon
dots and fullerenes. One-dimensional nanoparticles is in nano range in two dimen-
sions and out of nanoscale in the remaining one dimension; example, nanotubes and
nanowires [3]. In two-dimensional nanoparticles, they are nanoscale in one dimen-
sions and out of nano in two dimensions; for example, nanosheets and graphene.
Three-dimensional nanomaterials are composites or aggregates of different nano-
structures and are out of nanoscale in all the three dimensions.
Discovery of nanotechnology and its development is a milestone in cancer ther-
apy, especially in the diagnosis and drug delivery system, and it is an emerging
interdisciplinary branch. Nanoparticles are initially designed to carry small hydro-
phobic molecules, later which was also used for the delivery of antibodies, DNA,
peptides, imaging agents and other chemicals for treatments especially in the cancer
detection and treatment. In traditional drug delivery systems, oral ingestion and
intravascular injections are used for the chemo-distribution throughout the body by
the help of blood circulation systems. Efficiency of this systems are very less, since
a small part of the drugs is reached in the targeted area. Targeted drug delivery is
considered the main remedy to this problem, which helps in the complete delivery
of medicine in the targeted spot and reduces side effects [4].
Jatzkewitz was the first scientist in 1953 to develop a polymer-based drug deliv-
ery system. In 1960, Bangham et al. developed a liposomal-based drug delivery
system. Later in 1972, Scheffel et al. synthesized one micron sized human serum
albumin particles, which is used for human serum albumin micropore-based
albumin-bound paclitaxel (Abraxane) as drug delivery system [5, 6]. Liposomal
doxorubicin (Doxil) was the first Food and Drug Administration (FDA)-approved
nanodrug for the treatment of Kaposi’s sarcoma in AIDS patients [7]. In this method,
Doxil was delivered at tumour sites through passive targeting which is also used for
drug delivery in multiple myelomas and ovarian cancers [8]. Myocet and
DaunoXome are non-pegylated liposomal doxorubicin and former is approved by
Canada and European countries for the treatment of breast cancer and later was
approved by the FDA for the first-line treatment of advanced AIDS-related Kaposi’s
sarcoma. In 2012, FDA approved Marqibo (vincristine sulphate liposome injection)
for the treatment of Philadelphia chromosome negative (Ph− acute lymphoblastic
leukaemia (ALL) [9].
There are different types of cancer treatments using nanoparticles – antibody-
based targeting, aptamer-based targeting and ligand-based targeting. These mecha-
nisms are schematically represented in Fig. 1. Antibody-based targeting is a
well-established method for nuclear nanomedicine-based diagnostic and
Plant-Based Green Nanoparticles in Cancer Diagnosis and Chemotherapy 1389
Fig. 1 Schematic diagram of different mechanisms the cancer therapy on cells using nanoparticles
Gold nanoparticles are considered promising nanoparticles for imaging and treat-
ment owing to their ease of green synthesis and functionalization, biocompatibil-
ity, as well as non-toxicity. AuNPs are cancer in vivo therapist and diagnostic
agents. These nanoparticles are radiation enhancers, laser therapy enhancers, con-
trast agents and radiofrequency thermotherapy enhancers [10]. During the radio-
therapy treatment using AuNPs, higher dose was received by cancerous cells than
the normal cells. Gold is biocompatible, non-toxic nanoparticles with potential
biocompatibility and the LD50 of this material is about 3.2 g (Au) per kg of body
weight [11].
1390 A. John and R. E. Roy
AuNPs are wonderful candidates for sensing application in probing and imaging
tumour cells due to their ability to interact strongly with visible light. When exposed
to light, AuNP absorbs and scatters visible light because electrons in AuNP get
excited to a state of collective oscillation through surface plasmon resonance (SPR).
In the cancer imaging applications, AuNPs are targeted and accumulated in the
tumours and can be visualized in the region and cells under study through their
amazing optical scattering properties. Here, AuNPs can be detected by using phase
contrast optical microscopy, dark field microscopy, photothermal imaging, photo-
acoustic imaging, etc. Because of high atomic weight, AuNPs are used as a visual-
izing labelling agent and immunostaining at the ultrastructural level using
transmission electron microscopy [12]. It can be demonstrated that antibody-
conjugated AuNP can be located at the receptors present on the plasma membrane
of cells (Fig. 2).
Fig. 2 Schematic diagram for imaging cancer cells with receptors on the surface with the help of
antibody-conjugated gold nano particles
Plant-Based Green Nanoparticles in Cancer Diagnosis and Chemotherapy 1391
One of the main functions of AuNPs in the cancer treatment as a drug delivery
vehicle is in the selective delivery of medicines in the site. Sze, charge and surface
properties of the nanoparticles have affected the uptake of AuNPs into cells and
their subsequent intracellular fate. The nature of covalent/non-covalent interactions
in drug-AuNP conjugate structure and the means of drug release following intro-
duction of the drug-AuNP complexes to cells give an effective drug delivery strat-
egy. The usage of AuNP as a drug delivery agent is critical to monitor any toxic
effects of residual materials in the cell after delivery AND an ideal model is a bio-
degradable nanoparticle vector whose lifespan is limited to the therapeutic window
[13]. The non-toxic profile and biocompatibility make AuNP as an amazing drug
delivery vehicle in cells.
In the cancer treatment field, AuNPs are currently being used as AuNP of various
shapes and sizes are absorbed by cancer cells either through a ligand–receptor spe-
cific interaction or non-specific means. In actual practice, AuNPs are coupled with
drug molecule and an appropriate ligand which directed them only to tumour cells.
Conjugation of AuNP to polyethylene glycol (PEG), and the conjugation of AuNPs
with specific antibodies that bind unique biomarkers expressed on tumour cells are
two main methods described by Chen et al. [14]. Modified AuNPs being taken up
by cancer cells are shown in Fig. 3.
Fig. 3 Schematic diagram of drug delivery system, AuNPs conjugated with anti-cancer drugs and
ligands coupled with the receptors on the surface of tumour cells
1392 A. John and R. E. Roy
Conjugation with PEG modifies the properties of AuNPs, prevents AuNP aggre-
gation and lengthens their retention time in blood. This modification also facilitates
the specific accumulation of AuNPs in tumour cells over healthy cells since the
elevated permeability of poorly differentiated blood vessels around tumours follow-
ing angiogenesis as well as the decreased clearance rate caused by the deficit of
functional lymphatic vessels in tumours [15].
In cancer detection, AuNPs are focused on the study of its application as X-ray
contrast agents. Iodine-based compounds, currently used in computed tomography
(CT) contrast agents, have several limitations such as including short imaging times
due to rapid renal clearance, renal toxicity and the need for catheterization in many
cases. When compared with iodine compounds, AuNPs are permitting longer imag-
ing time since they stay in the blood for a longer time than iodine agents.
Tissue distinguishability between different types of tissues based on computed
thermography (CT) depends on difference in degree of X-ray attenuation, where
attenuation coefficient is a function of the atomic number and electron density of the
contrast agents. AuNPs have more advantages over iodine-based contrast agents
because the atomic number and electron density of gold (79 and 19.32 g/cm3, respec-
tively) are much higher than those of the currently used iodine (53 and 4.9 g/cm3).
Therefore, when compared with the iodine compounds, AuNPs have higher absorp-
tion and better contrast can be achieved with lower X-ray dose. AuNPs are also used
for the enhanced detection of blood vessels. In 1970s, Dr. Judah Folkman of the
Harvard Medical School suggested that the cancer growth and metastasis depend on
angiogenesis for nutrients and oxygen and we can fight against cancer by inhibiting
new blood vessel formation [16]. In an advanced study, Chien et al. injected AuNPs
(1.9 nm in diameter) intravenously and recorded images over time with a standard
mammography unit. In this study, cancer tumours and kidney were seen with unusual
clarity and high spatial resolution. AuNPs are highly efficient and the smallest mea-
sured blood vessel lumen diameters were 3–5 μm. Computed thermography contrast
agent can be developed using antibiofouling polymer-coated AuNPs, gadolinium-
coated AuNPs, PEG-coated nanoparticles and antibody-coated nanoparticles.
biochemical uses. AuNPs can be easily synthesized using AuNPs using common
stabilizing agents such as sodium citrate, transferrin and cetyltrimethylammonium
bromide, while common species used for functionalization include various amines,
oligonucleotides, peptides, antibodies and lipids. Selection of different plants, algae,
bacteria and fungi as reducing agents and stabilizing agents for the size selective
biocompatible gold nanoparticle synthesis is a highly studied area [17]. For exam-
ple, AuNPs with particle-size distributions in the range of 2–12 nm were synthe-
sized by Raveendran et al. using glucose as a reducing agent and starch as a
protective agent. Huang et al. used Bayberry’s tannin extract simultaneously as
reducing and protective agents for the synthesize of AuNPs with an average size of
2 nm. Sharma et al. reported a synthetic root for AuNP preparation from the tea leaf
extract. Different concentration of the tea leaf extract could be used to control the
size of synthesized nanoparticles. Suman et al. [18] used extract from Morinda citri-
folia roots at room temperature for the synthesize of 8–17 nm sized AuNPs.
10 g of leaf, fruit or any other plant part is weighed and washed thoroughly four
times using double distilled water. This clean plant parts were cut into small pieces
and put into a blade blender with 30 mL of double distilled water until a homoge-
neous fluid was obtained. The plant extract was then diluted to 50 mL using distilled
water and larger particles in the extract can be removed through centrifugation for
2 min, followed by filtering through a 0.45 μm filter and the extracts were kept fro-
zen at −18 °C until used. In the green method of AuNP synthesis, consumable plant
parts with high antioxidant content were employed to ensure biocompatibility.
AuNPs could be synthesized by mixing 10 mL of the gold ion solution (1.0 mg
HAuCl4) with 0.75 mL of plant extract. Deep purple/red solution will be formed
within a few seconds, which is the indication of AuNP formation [19]. By changing
the time, temperature and pH, AuNP with varying size and shape can be synthesized
from plant extract (Fig. 4).
Fig. 4 Different steps in the synthesis of gold nanoparticle using plant extract
1394 A. John and R. E. Roy
In recent years, nanoparticles of relatively noble metals such as gold, silver, copper
and palladium have drawn immense attention due to the wide range of new applica-
tions in various interdisciplinary areas including biochemical, medicinal, industrial
fields. Particularly, silver nanoparticles (AgNPs) have significant interest in medical
applications such as cancer imaging, cancer treatment, anti-bacterial agents without
toxic effects and industrial applications such as inkjet links containing well uniform
dispersions of nanosized silver particles that are useful for producing electronic
circuits [20]. For the proper utilization of AgNP for various applications, it requires
that the particles must be of nanosize and they must be produced easily as well as at
low cost and time consumption (Fig. 5).
In the ancient years, silver was used as an anti-inflammatory and anti-bacterial agent.
The most widely used silver compounds in biomedical application is silver sulphadi-
azine that is now replaced by the nanosilver. Green-synthesized AgNP is used in
Fig. 5 Green synthetic route for silver nanoparticle synthesis from plant extract
Plant-Based Green Nanoparticles in Cancer Diagnosis and Chemotherapy 1395
Biogenic synthesis of AgNPs is known for many researchers from long time.
Biogenic synthetic routes have several advantages over traditional chemical or
physical ones, since they are eco-friendly. Biological approach for synthesizing
AgNPs is being increasingly considered over the past few decades. This method is
a green technology aimed at minimizing the negative environmental impact. It had
been known that the synthesis of AgNPs using the chemical approach requires three
main ingredients; a silver salt, a reducing agent and the stabilizer which are replaced
using molecules obtained from living organisms such as plants, bacteria, fungi,
yeast and algae in biological approach [27, 28]. Plant extracts are eco-friendly
reducing agents used for the synthesis of AgNPs and are discussed below:
1396 A. John and R. E. Roy
20 g of fresh leaves or plant parts were collected and washed with distilled water
to remove all the unwanted dust particles, dried and the leaves were cut into small
pieces. The weighed leaves were transferred into a clean beaker containing 200 mL
of distilled water. It is boiled for 30 min. Then the extract was cooled and filtered
using Whatmann No. 1 and stored at 4 °C.
5 mL of plant extract was then added to 10 mL of 1 M (0.016 g) AgNO3 solution.
Shake well and keep the extract for 24 h in dark condition. The colour changes from
pale green to reddish brown indicating the formation of AgNPs.
Fig. 6 Steps in silver nanoparticle synthesis and characterization using plant extract
1398 A. John and R. E. Roy
Fig. 7 Green synthetic route for the synthesis of green copper nanoparticle from plant extract
Polymer-based nanoparticles are emerging systems that are used in preclinical stud-
ies and as promising delivery agents. Polymers serve as a structural engineer for the
preparation of multifunctional nanoparticles with suitable size, shape and surface
functionalization. The excellent pharmacokinetic properties of polymeric nanopar-
ticles make them an ideal candidate for drug delivery applications. It is reported that
for the initiation, propagation and regeneration of cancers, the cancer stem cells
(CSCs) play a major role. Hence, recent studies report the functionalization of CSCs
on to the exterior or interior of the PNPs for the successful elimination of CSCs.
Polymeric drug-loaded nanoparticles are the best delivery systems developed so
far because of their excellent pharmacokinetic behaviour, size of particles, rate of
drug loading, surface charge and chemistry, speed of degradation etc., need to be
included [35–37]. Surface functionalization of PNPs makes the suitable as an effi-
cient drug vehicle by effectively accumulating the loaded drugs in cancer cells with-
out harming the healthy cells [36].
Some of the polymers in recent research, selectively used for CSC therapy,
include poly (D, L-lactic-co-glycolic acid) (PLGA), polylactic acid (PLA), poly
(ethylene glycol) (PEG), chitosan (CS) and hyaluronic acid (HA) [37].
Plant-Based Green Nanoparticles in Cancer Diagnosis and Chemotherapy 1399
The highly hydrophilic PEG with exceptional solubility, free mobility, non-toxicity
and non-immunogenicity finds a wide application in the biomedical field. Shen
et al. used poly(ethylene glycol)-block-poly(d,l-lactide) (PEG-b-PLA) nanoparti-
cles into highly non-polar botezomib used in clinical applications. The drug-loaded
PEG-b-PLA nanoparticles selectively destroyed CSCs thus increasing the applica-
bility of bortezomib in breast cancer therapy [46]. Studies were also carried on
diblock copolymer nanoparticles of PEG and surface modified polycarbonates
(PAC) synthesized using ring opening polymerization which improved the efficacy
1400 A. John and R. E. Roy
The biodegradable polymer, PLA has been effectively used as a polymeric drug-
loaded nanoparticle delivery system in the treatment of lung cancer stem cells [48].
The PLA nanoparticles were embedded in docetaxel (DTX) and the results proved
remarkable anti-metastatic act of this dug-loaded nanoparticle system in both in
vitro and in vivo experiments. A similar drug-loaded nanoparticle system based on
PLA nanoparticles was also developed [49] using the method of emulsion solvent
evaporation which could effectively inhibit the growth of tumours and also improved
its anti-metastatic capability in the treatment of cancer [50]. Another drug delivery
system based on PLA nanoparticles encapsulated in quercetin (Qt) was developed
by Pandey et al. for the treatment of breast cancer cells [51]. A new system of PLA
nanoparticles grafted on to cellulose was developed to act on betulinic drug [CE-g-
PLL/BA] which could successfully suppress the growth of tumours with few side
effects than BA alone [52].
5.5 Chitosan
6 Conclusion
This chapter gives a comprehensive idea about activated and regulated molecular
pathways of different green-synthesized nanoparticles that are causing cancer cell
imaging and death. Optimal drug delivery strategy is based on receptor-based active
selective targeting of nanoparticles. Tumour-based nano-delivery vehicles are
employed for early cancer detection, therapy and post-therapeutic follow-up. Mode
of action and intensity of different green-synthesized nanoparticles vary from one
form of cancer to the other, which depends on the nanoparticle material, size and
shape of the particle, selection of plant precursor etc. Apart from the properties of
green plant-mediated nanoparticles, the cellular response is also crucial. ROSs can
be considered as a precursor molecule of cell death because they are the initiator
molecule in apoptotic, autophagic and necroptotic pathways. In nano-therapy, the
main objective is to utilize nanoparticles for the in vivo targeted selective killing of
cancer cells with no or minimal side effects.
This area is an emerging research field, in which the development of effective,
less toxic, biocompatible, selective drug delivery systems is most important in the
cancer cell identification and treatment. The potential of green-synthesized plant
genic nanoparticle in therapeutic in vitro systems has been explored, while 3D
tumour model studies and clinical trials remain unexplored.
References
1. Khanna SC, Speiser P (1969) Epoxy resin beads as a pharmaceutical dosage form I: methods
of preparation. J Pharm 58:1114–1117
2. Speiser P, Khanna SC (1970) Perlpolymerisate, eine neue perorale Darreichungsform und ihre
Beeinflussung durch Arzneistoffe. Prapar Pharm 6:1–4
3. Hasan S (2015) A review on nanoparticles : their synthesis and types biosynthesis mechanism.
Res J Recent Sci 4:9–11
4. Korbekandi H, Iravani S (2012) In: Hashim AA (ed) Silver nanoparticles, the delivery of
nanoparticles; ISBN: 978-953-51-0615-9.
5. Machado S, Pacheco JG, Nouws HPA, Albergaria JT, Delerue-Matos C (2015) Characterization
of green zero-valent iron nanoparticles produced with tree leaf extracts. Sci Total Environ
533:76–81
6. Kreuter J (1983) Evaluation of nanoparticles as drug delivery systems I: preparation methods.
Pharm Acta Helv 58:196–209
7. Kommareddy S, Tiwari SB, Amiji MM (2005) Long-circulating polymeric nanovectors for
tumor-selective gene delivery. Technol Cancer Res Treat 4:615–625
8. El-Sayed MA (2001) Some interesting properties of metals confined in time and nanometer
space of different shapes. Acc Chem Res 34(4):257–264
9. Link S, El-Sayed MA (1999) Spectral properties and relaxation dynamics of surface plas-
mon electronic oscillations in gold and silver nanodots and nanorods. J Phys Chem B
103(40):8410–8426
10. Link S, El-Sayed MA (2000) Shape and size dependence of radiative, non-radiative and pho-
tothermal properties of gold nanocrystals. Int Rev Phys Chem 19(3):409–453
11. Weissleder R (2001) A clearer vision for in vivo imaging. Nat Biotechnol 19(4):316–317
1402 A. John and R. E. Roy
12. Huang XH, El-Sayed IH, Qian W, El-Sayed MA (2006) Cancer cell imaging and photothermal
therapy in the near-infrared region by using gold nanorods. J Am Chem Soc 128(6):2115–2120
13. Boisselier E, Astruc D (2009) Gold nanoparticles in nanomedicine: preparations, imaging,
diagnostics, therapies and toxicity. Chem Soc Rev 38:1759–1782
14. Chen YS, Hung YC, Liau I, Huang GS (2009) Assessment of the in vivo toxicity of gold
nanoparticles. Nanoscale Res Lett 4:858–864
15. Cho W, Cho M, Jeong J, Choi M, Cho H, Han B et al (2009) Acute toxicity and pharmacokinet-
ics of 13 nm-sized PEG-coated gold nanoparticles. Toxicol Appl Pharmacol 236:16–24
16. Kim C, Ghosh P, Rotello V (2009) Multimodal drug delivery using gold nanoparticles.
Nanoscale 1:61–67
17. Kumar KM, Mandal BK, Sinha M, Krishnakumar V (2012) Terminalia chebula mediated
green and rapid synthesis of gold nanoparticles. Spectrochim Acta A Mol Biomol Spectrosc
86:490–494
18. Suman TY, Radhika Rajasree SR, Ramkumar R, Rajthilak C, Perumal P (2014) The green syn-
thesis of gold nanoparticles using an aqueous root extract of Morinda citrifolia L. Spectrochim
Acta A Mol Biomol Spectrosc 118:11–16
19. Shankar SS, Ahmad A, Pasricha R, Sastry M (2003) Bioreduction of chloroaurate ions by
geranium leaves and its endophytic fungus yields gold nanoparticles of different shapes. J
Mater Chem 13(7):1822–1826
20. He Y, Du Z, Ma S, Cheng S, Jiang S, Liu Y, Li D, Huang H, Zhang K, Zheng X (2016)
Biosynthesis, antibacterial activity and anticancer effects against prostate cancer (PC-3) cells
of silver NPs using Dimocarpus Longan Lour. peel extract. Nanoscale Res Lett 11(1):300
21. Priyadharshini RI, Prasannaraj G, Geetha N, Venkatachalam P (2014) Microwave-mediated
extracellular synthesis of metallic silver and zinc oxide NPs using macro-algae (Gracilaria
edulis) extracts and its anticancer activity against human PC3 cell lines. Appl Biochem
Biotechnol 174(8):2777–2790
22. Netala VR, Bethu MS, Pushpalatha B, Baki VB, Aishwarya S, Rao JV, Tartte V (2016)
Biogenesis of silver NPs using endophytic fungus Pestalotiopsis microspora and evaluation of
their antioxidant and anticancer activities. Int J Nanomedicine 11:5683
23. Padinjarathil H, Joseph MM, Unnikrishnan BS, Preethi GU, Shiji R, Archana MG, Maya
S, Syama HP, Sreelekha TT (2018) Galactomannan endowed biogenic silver NPs exposed
enhanced cancer cytotoxicity with excellent biocompatibility. Int J Biol Macromol
118:1174–1182
24. Farah MA, Ali MA, Chen SM, Li Y, Al-Hemaid FM, Abou-Tarboush FM, Al-Anazi KM,
Lee J (2016) Silver NPs synthesized from Adenium obesum leaf extract induced DNA dam-
age, apoptosis and autophagy via generation of reactive oxygen species. Colloids Surf B
Biointerfaces 141:158–169
25. Remya RR, Rajasree SR, Aranganathan L, Suman TY (2015) An investigation on cytotoxic
effect of bioactive AgNPs synthesized using Cassia fistula flower extract on breast cancer cell
MCF-7. Biotechnol Rep 8:110–115
26. Jeyaraj M, Sathishkumar G, Sivanandhan G, Mubarak Ali D, Rajesh M, Arun R, Kapildev G,
Manickavasagam M, Thajuddin N, Premkumar K, Ganapathi A (2013) Biogenic silver NPs for
cancer treatment: an experimental report. Colloids Surf B Biointerfaces 106:86–92
27. Jiang H, Manolache S, Wong ACL, Denes FS (2004) Plasma enhanced deposition of silver
nanoparticles onto polymer and metal surfaces for the generation of antimicrobial characteris-
tics. J Appl Polym Sci 93:1411–1422
28. Chandran SP, Chaudhary M, Pasricha R, Ahmad A, Sastry M (2006) Synthesis of gold nano-
triangles and silver nanoparticles using Aloe vera plant extract. Biotechnol Prog 22:577–583
29. Krishnaraj C, Muthukumaran P, Ramachandran R, Balakumaran MD, Kalaichelvn PT (2014)
Acalypha indica Linn: biogenic synthesis of silver and gold NPs and their cytotoxic effects
against MDA-MB-231, human breast cancer cells. Biotechnol Rep 4:42–49
Plant-Based Green Nanoparticles in Cancer Diagnosis and Chemotherapy 1403
30. Nagajyothi PC, Muthuraman P, Sreekanth TV, Kim DH, Shim J (2015) Green synthesis:
in vitro anticancer activity of copper oxide NPs against human cervical carcinoma cells. Arab
J Chem 10(2):215–225
31. Shirisha R, Varalakshmi KN (2017) Tamarindus indica bark extract and its bioactive fraction
induce apoptosis in HeLa and PA-1 cells. Indian J Pharm Sci 78(6):725–731
32. Sulaiman GM, Tawfeeq AT, Jaaffer MD (2018) Biogenic synthesis of copper oxide NPs using
olea europaea leaf extract and evaluation of their toxicity activities: an in vivo and in vitro
study. Biotechnol Prog 34(1):218–230
33. Abboud Y, Saffaj T, Chagraoui A, El Bouari A, Brouzi K, Tanane O, Ihssane B (2014)
Biosynthesis, characterization and antimicrobial activity of copper oxide nanoparticles
(CONPs) produced using brown alga extract (Bifurcaria bifurcata). Appl Nanosci 4:571–576
34. Angrasan JKVM, Subbaiya R (2014) Biosynthesis of copper nanoparticles by Vitis vinifera
leaf aqueous extract and its antibacterial activity. Int J Curr Microbiol App Sci 3(9):768–774
35. Vinogradov S, Wei X (2012) Cancer stem cells and drug resistance: the potential of nanomedi-
cine. Nanomedicine 7:597–615. https://doi.org/10.2217/nnm.12.22
36. Fonseca AC, Ferreira P, Cordeiro RA, Mendonça PV, Góis JR, Gil MH et al (2013) Drug
delivery systems for predictive medicine: polymers as tools for advanced applications. In:
Mozaffari MS (ed) New strategies to advance pre/diabetes care: integrative approach by
PPPM. Springer, Dordrecht
37. Fonseca AC, Serra AC, Coelho JFJ (2014) Bioabsorbable polymers in cancer therapy: latest
developments. EPMA J 6:1–18. https://doi.org/10.1186/s13167-015-0045-z
38. Chaubal M (2002) Polylactides/glycolides-excipients for injectable drug delivery and beyond.
Drug Deliv Technol 2:34–36
39. Astete CE, Sabliov CM (2006) Synthesis and characterization of plga nanoparticles. J Biomater
Sci Polym Ed 17:247–289. https://doi.org/10.1163/156856206775997322
40. Biondi M, Ungaro F, Quaglia F, Netti PA (2008) Controlled drug delivery in tissue engineer-
ing. Adv Drug Deliv Rev 60:229–242. https://doi.org/10.1016/j.addr.2007.08.038
41. Muntimadugu E, Kumar R, Saladi S, Rafeeqi TA, Khan W (2016) Cd44targeted chemother-
apy for co-eradication of breast cancer stem cells and cancercells using polymeric nanopar-
ticles of salinomycin and paclitaxel. Colloids Surf B Biointerfaces 143:532–546. https://doi.
org/10.1016/j.colsurfb.2016.03.075
42. Luo Y, Ziebell MR, Prestwich GD (2000) A hyaluronic acid-taxolantitumor bioconjugate tar-
geted to cancer cells. Biomacromolecules 1:208–218. https://doi.org/10.1021/bm000283n
43. Prestwich GD (2011) Hyaluronic acid-based clinical biomaterials derived for cell and molecule
delivery in regenerative medicine. J Control Release 155:193–199. https://doi.org/10.1016/j.
jconrel.2011.04.007
44. Banzato A, Bobisse S, Rondina M, Renier D, Bettella F, Esposito G et al (2008) A paclitaxel-
hyaluronan bioconjugate targeting ovarian cancer affords a potent in vivo therapeutic activity.
Clin Cancer Res 14:3598–3606. https://doi.org/10.1158/1078-0432.CCR-07-2019
45. Choi KY, Chung H, Min KH, Hong YY, Kim K, Park JH et al (2009) Self-assembled hyal-
uronic acid nanoparticles for active tumor targeting. Biomaterials 31:106–114. https://doi.
org/10.1016/j.biomaterials.2009.09.030
46. Choi KY, Min KH, Yoon HY, Kim K, Park JH, Kwon IC et al (2011) PEGylation of hyaluronic
acid nanoparticles improves tumor targetability in vivo. Biomaterials 32:1880–1889. https://
doi.org/10.1016/j.biomaterials.2010.11.010
47. Shen S, Du XJ, Liu J, Sun R, Zhu YH, Wang J (2015) Delivery of bortezomib with nanopar-
ticles for basal-like triple-negative breast cancer therapy. J Control Release 208:14–24. https://
doi.org/10.1016/j.jconrel.2014.12.043
48. Ke XY, Ng VWL, Gao SJ, Tong YW, Hedrick JL, Yang YY (2014) Co-delivery of thioridazine
and doxorubicin using polymeric micelles for targeting both cancer cells and cancer stem cells.
Biomaterials 35:1096–1108. https://doi.org/10.1016/j.biomaterials.2013.10.049
1404 A. John and R. E. Roy
49. Yang N, Jiang Y, Zhang H, Sun B, Hou C, Zheng J et al (2015) Active targeting docetaxel-PLA
nanoparticles eradicate circulating lung cancer stem like cells and inhibit liver metastasis. Mol
Pharm 12:232–239. https://doi.org/10.1021/mp500568z
50. Liang Z, Yang N, Jiang Y, Hou C, Zheng J, Shi J et al (2015) Targeting docetaxel-pla nanopar-
ticles simultaneously inhibit tumor growth and liver metastases of small cell lung cancer. Int J
Pharm 494:337–345. https://doi.org/10.1016/j.ijpharm.2015.08.042
51. Pandey SK, Patel DK, Thakur R, Mishra DP, Maiti P, Haldar C (2015) Anti-cancer evaluation
of quercetin embedded pla nanoparticles synthesized by emulsified nanoprecipitation. Int J
Biol Macromol 75:521–529. https://doi.org/10.1016/j.ijbiomac.2015.02.011
52. Dai L, Yang T, He J, Deng L, Liu J, Wang L et al (2014) Cellulosegraft- poly(l-lactic acid)
nanoparticles for efficient delivery of anti-cancer drugs. J Mater Chem B 2:6749–6757. https://
doi.org/10.1039/C4TB00956H
53. Senel S, Mcclure SJ (2004) Potential applications of chitosan in veterinary medicine. Adv
Drug Deliv Rev 56:1467–1480. https://doi.org/10.1016/j.addr
54. Nascimento AV, Singh A, Bousbaa H, Ferreira D, Sarmento B, Amiji MM (2015)
Combinatorial-designed epidermal growth factor receptortargeted chitosan nanoparticles for
encapsulation and delivery of lipid-modified platinum derivatives in wild-type and resistant
non-small-cell lung cancer cells. Mol Pharm 12:4466–4477. https://doi.org/10.1021/acs.
molpharmaceut.5b00642
55. Rao W, Wang H, Han J, Zhao S, Dumbleton J, Agarwal P et al (2015) Chitosan-decorated
doxorubicin-encapsulated nanoparticle targets and eliminates tumor reinitiating cancer stem-
like cells. ACS Nano 9:5725–5740. https://doi.org/10.1021/nn506928p
Use of Plant-Derived Nanoparticles
in Cancer Therapy
leguminous plants, which include soybeans, lupine and fava beans. Studies show
that genistein exhibits a number of beneficial biological effects: heart disease, dia-
betes, neuropathy, osteoporosis, inflammatory diseases and cancer. In the last
decade, a few cancer-related clinical trials on genistein, including colorectal, pros-
tate, pancreatic, breast and kidney cancer, have been diagnosed worldwide.
Meanwhile, the inhibition of the effects of genistein on different types of cancer and
subcutaneous pathways are under investigation. Like phytoestrogen, genistein can
competitively bind to oestrogen receptors (ERs) and affects oestrogen-dependent
cancer (Fig. 1).
Hwang et al, reported that genistein inhibits the growth of ovarian cancer cells
(BG-1) by suppressing both ER and insulin-like growth factor-1 receptor (IGF-1R)
signalling methods [3]. Genistein also presses proliferation and secretion of MCF-7
and 3T3-L1 breast cancer cells by reducing regulation of ERα expression [4]. In
addition, genistein induces apoptosis by inhibiting the nuclear factor-kappa B (NF-
κB) pathway in LoVo and HT-29 human colon cancer cells [5] and T-cell leukaemia
Nanotechnology has been shown to help the success of cancer drug development.
Several phytochemical nanoparticles are commercially available. Compared to free
phytochemicals, phytochemical nanoparticles show not only an increase in drug
solubility but also other benefits.
Changes in pharmacokinetics and distribution of phytochemicals due to the com-
position of nanoparticles can lead to improving the therapeutic index of these
Use of Plant-Derived Nanoparticles in Cancer Therapy 1409
allows them to focus on the area of cancer cells. The effect of EPR is not present in
normal muscles. The most important factors that influence the impact of EPR were
the size and consistency of biocompatibility. The minimum size of the EPR result is
cell size greater than 40 kDa (macromolecules) or particle size greater than 5 nm
(nanocarrier) (Fig. 3) [20]. It takes a lot of rotation time to give enough time for
delivery of the drug through the EPR effect. Many phytochemicals are low-weight
agents with rapid approval in vivo and widespread distribution in normal organs and
tissues [21]. MDR is one of the main causes of the failure of phytochemicals in
cancer. Using nanovehicles, the delivery of phytochemicals is a new strategy to
overcome MDR.
Modification of facial nanocarriers can improve the delivery of phytochemicals
and defeat drug resistance by altering biophysical interactions between nanocarriers
and cancer cell membrane lipids and increase the delivery of phytochemicals to
target tissues [22]. Chemical modification of phytochemicals and recruitment of
drug delivery systems are widely used for the delivery of phytochemicals. For chem-
ical repair, we focus on selected drug candidates who have been tested in clinical
trials or who have the ability to enter the clinic temptations. With the delivery pro-
grams of the phytochemical component, we will focus on the former FDA approved
(Fig. 4).
3 Nanoparticles
cancer were performed at M. D. Anderson Cancer Centre, Texas, USA. This study
focuses on the toxicity profile, magnitude tolerated dose (MTD), as well as the
pharmacokinetics properties of Abraxane. The schedule was administered every
3 weeks using Abraxane dosage pump from 135 to 375 mg/m2. The results showed
that Abraxane was well tolerated, producing only neuropathy in three patients, sto-
matitis in two patients, and external keratopathy in two patients with doses of
1412 R. S. Nandhini et al.
ABI-007 at 375 mg/m2. They concluded that the Mett of Abraxane was 300 mg/m2.
The plasma AUC of Abraxane was higher than that of Taxol if 260 mg/m2 of
Abraxane was injected for 30 min i.e., 175 mg/m2 of Taxol injected as 3 h. Abraxane
provided an example of albumin nanoparticles that will be used in clinical practice
[31, 32]. Currently, Abraxane progresses to Phase II/III clinical trials for metastatic
breast cancer patients with i.e., dose at 260 mg/m2 in 3-week cycles compared to the
standard paclitaxel system, taken at 175 mg/m2. Compared to Taxol, Abraxane indi-
cated that patients’ response rates were significantly higher (19% vs 33%, respec-
tively) and much longer length of plant growth period (16.9 weeks vs 23.0,
respectively) [32, 33].
3.1 Silver Nanoparticles
Plant-based silver nanoparticles have been repeatedly tested against various human
cancer cell lines like breast cells, liver cells and colon cells. While compared to
other routine physical and chemical approaches in the treatment of this lethal dis-
ease, green synthesis of silver nanoparticles by implementing the abilities of various
secondary metabolites from plants has been proven to be more efficient, ecofriendly,
cost-friendly and safer [34].
For example, using the bark and leaf extracts from the tree Ziziphus xylopyrus
which is very common in the Indian sub-continent, biofabrication of silver nanopar-
ticles (AgNPs) was possible. Not only the Zizipus tree but also several other com-
mon plants and trees like hibiscus, polyalthia, papaya, Ocimum, Coriandrum and
Ficus are employed in the biosynthesis of silver nanoparticles [35].
Green synthesis of nanoparticles over synthetic nanoparticles has paved way to
overcome some serious problems like aggregation of nanoparticles, toxicity and
non-compatibility, which offers advantages like stability, faster reduction rate
and so on.
Benefits of
using green
nanoparticles
dna, reduced
reduced improved faster improved
mitrochondrial compatibility
toxicity stability reduction rate apoptosis
damage complications
Many research studies have proved that silver nanoparticles have beneficial toxic
mechanisms, which are capable of causing DNA and mitochondrial damage and
inducing apoptosis (programmed cell death) in cancer cells. For instance, green
synthesis of the plant Datura inoxia, activities like cell cycle arrest, decreased DNA
synthesis and apoptosis were recorded in cancer cells [36].
Use of Plant-Derived Nanoparticles in Cancer Therapy 1413
Interestingly enough, not just terrestrial plants but even marine corals were
reported to show anti-cancer properties when used to synthesise silver nanoparti-
cles. For example, extracts from the soft coral Cladiella pachyclados were used in
producing bioactive silver nanoparticles. This coral possesses a compound called
eunicellin, a diterpenoid that has a great potential against breast cancer [37].
This compound, when used to synthesise AgNPs or using silver nanoparticles as
drug carriers, showed anti-inflammatory potential almost equal to the commonly
used cancer drug Doxorubicin; in fact, it was able to express high selectivity
comparatively.
The experiment showed that silver nanoparticles synthesised using the extracts
from the mint plant as a stabilising agent exhibited a significant potential against the
HCT116 human cancer cell line [36]. This analysis showed that plant-mediated
AgNPs were capable of suspending cell division and minimising the further prolif-
eration of cancer cells.
AgNPs have proved themselves very useful in the manufacture of nanodrugs.
They use a mechanobiological and microenvironmental mechanism to cause apop-
totic cell death in carcinogenic cells with the aid of ROS-mediated pathways. For
example the chemotherapeutic drug—fructose coated angstrom silver has been
reported to inhibit osteosarcoma cells (a malignant bone cancer), pancreatic and
lung cancer cells. Further experiments showed the drug to be capable of inhibiting
tumour growth, improving survival rate and attenuating osteolysis in osteosarcoma-
bearing mice [38].
Therefore, green-synthesised silver nanoparticles are being extensively studied
by researchers worldwide to be implemented in the cure of cancer effective imme-
diately, which could be a significant breakthrough, given its properties and eco-
friendly nature.
3.2 Nanodiamonds
breast cancer cells in mice, the nanodiamond mediated doxorubicin delivery system
was well able to cease the generation of lung metastasis [43]. Thus, with the help of
nanodiamonds, the drug was able to be successfully delivered in several lab tri-
als which the researchers have speculated using Nano diamonds as intercellular
carriers of plant metabolites, like the plant metabolites quercetin and citropten are
intensely being studied for their pro-apoptotic and redox properties [42].
Quercetin is a natural pigment (flavonoid) that is available in most common
fruits, grains and vegetables and is nowadays taken as a dietary supplement. Due to
its high level of free radicals, the flavonoid promotes inflammation in human cells
which is a very important anti- cancer activity.
Citropten or 5,7-dimethoxycoumarin is a metabolite (coumarin) extracted from
a plant called Peleaanisata and several natural oils and citrus fruits which is known
for inducing melanogenesis [44].
Extensive studies have been carried out to assess whether these plant metabolites
were able to show improved antiproliferative and inflammatory effects when
adsorbed on nanodiamonds.
It was speculated that the bioactivities of these plant metabolites were modifiable
through their interaction with nanodiamonds.
When tests were performed on the HeLa cell line, it was evident that the conju-
gates were able to first penetrate the cell compartment, next, the intracellular spaces,
following the areas near the nuclear region successfully.
On the whole, based on the experiments performed on the HeLa and B16F10 cell
lines, it was clear that the nanodiamonds heightened the cell growth inhibitory prop-
erties of the two plant metabolites quercetin and citropten, making it an effective
agent against cancer [39].
3.2.1
In Silico Studies of Plant Nanoparticles in Cancer Therapy
In silico studies are a very crucial step in the process of drug design. These are
computer-aided analyses that are carried out in order to know several properties like
the binding affinity which are very important in order to design an approvable drug.
4 Liposomes
5 Micelle
study, Genexol-PM showed strong inhibition of tumour growth and higher MTD
than-Taxol®. A few of the Genexol-PM clinical trials have been in operation or in
place completed, with indications including non- small cell lung cancer, metastatic
breast cancer and pancreatic cancer [55]. In the first phase of clinical trials,
Genexol-PM showed MTD 2.3 times higher than Taxol® in humans [56]. In addi-
tional clinical research, Genexol-PM has shown better performance than Taxol
when used alone and as part of a combined treatment. However, it is interesting that
Genexol-PM showed a very low T 1/2 (12 h vs 20 h) and AUC 11.58 compared to
Taxol®, possibly due to Genexol-PM faster approval and better access to cancer.
6
Cladiella pachyclados
Out of the 26 compounds that were tested with the aid of inverse molecular dock-
ing, only six were found to show binding orientations inside the corresponding
binding sites. They also were reported to have the highest dG values, proving that
they could be used as potential binders for several BC proteins [37].
7 Flavonoids
As we discussed the usage of flavonoids like the quercetin in the treatment of can-
cer, there are many other such flavonoids which possess beneficial anti-cancer prop-
erties. For example, MYRICETIN is one of the plants derived flavonoids which
contains a lot of strong anticancer properties. This compound that is obtained from
common plants and fruits is also one of the major constituents of several food and
beverages [58]. Myricetin is proven to exhibit cytotoxic activity towards a huge
number of human cancer cell lines.
Research studies show that the biosynthesis of Myr-Au NPs resulted in spherical-
shaped particles of size less than 50 nm [59].
mTOR is the mammalian target of rapamycin which is a kinase molecule that
regulates protein synthesis. A theoretical network analysis was carried out by the
Kyoto Encyclopedia of Genes and Genomes (KEGG) database with the help of a
software called Cytoscape 3.7.1 to understand the role of different genes in the
mTOR signalling mechanism. The observation of the said analysis is shown below
[59] (Table 1).
Molecular docking of myricetin (Myr) guided in understanding the X-ray crystal
structure of human mTOR kinase enzyme obtained from the RCSB-PDB (Research
Collaboratory for Structural Bioinformatics-Protein Data Bank).
The GLIDE program (v.11, Schrödinger, LLC, New York, 2016) was utilised in
extra precision (XP) docking mode with the Glidescore and E-model scoring func-
tions to perform the molecular docking analysis of myricetin for the mTOR gene.
MTT assay that is a colorimetric assay performed for assessing the cell meta-
bolic activity was done, and ROS generation studies were carried out and the values
were compared using SPSS (v. 20.0, SPSS Inc., Chicago, IL, USA).
In this analysis of Myr-AuNPs, extra precision GLIDE docking procedure was
done by removing the inhibitor compound with the human mTOR receptor. The
docking result of myrecitin was found to be −7.79 kcal mol−1, and its interactions
were confirmed with ALA 1708, ARG 1709, LYS 1710, ILE 1711 and ASP 1712 of
chain B, thereby proving a good binding activity of Myrecitin with human mTOR
kinase. The molecular docking studies in mTOR kinase were understood to show a
strong binding affinity with IC50 value = 13 μg mL–1. After staining, the cells treated
with myr-AuNPs showed a good proportion of dead cells, thereby showing an
anticancer activity [59].
8 Chitosan
9 Pro-haloacetate NPs
reversing the mitochondrial suppression with metabolic modulating drugs like PDK
inhibitors. Recent molecular modelling studies show that the haloacetic acids are
capable of selective recognition of certain PDK isoforms allowing us to recognise
better haloacetate drug candidates. The quantitative docking energy data from the
molecular recognition analysis indicated that MCA (mono chloroacetate) was a
comparable PDK binder while DBA (di bromo acetate) showed a slightly weaker
binding than the DCA [49]. For further process of molecular modelling, the above-
mentioned haloacetates were prepared with the help of SKETCH module. The fol-
lowing requirements was set up to carry out the procedure, followed by the
results [49].
With the growing necessity day by day to develop environment and economy-
friendly approaches in the cure of the deadly disease cancer, the utilisation of plant
nanoparticles has indeed offered a huge helping hand in this deed, given its numer-
ous advantages over several other existing techniques.
Employment of plants in the field of medicine especially in curing diseases is not
new in our civilisation. Taking this under consideration, the aid of green synthesis
of nanoparticles or simply green nanoparticles has improved not just the methods
but, ecofriendly and very achievable methods of treating cancer in multitudes.
Nowadays, synthesising nanoparticles with the aid of plants is a majorly dis-
cussed topic due to their unbelievably advantageous properties and safe nature.
Using commonly available plants, several nanoparticles have been synthesised suc-
cessfully. Few examples are given in Tables 2 and 3 [97].
1420 R. S. Nandhini et al.
Table 2 List of various common plants from where different nanoparticles have been synthesised
Plant Nanoparticle synthesised Size (nm)
Aloe vera Au & Ag 50–350
Curcuma longa Pd 10–15
Eucalyptus macrocarpa Au Ag 20–100
10–100
Mangifera indica Ag 20
Psidium guajava Au 25–30
Caria papaya Ag 15
Citrus sinensis Ag 35
Following this, further researches theorised that the use of nanosystems can
reduce those toxic side effects by a great proportion with the help of Fucoidan, a
polysaccharide extracted from brown algae and certain marine invertebrates [102].
This compound is used widely in the green synthesis of nanoparticles using AEP
method aiding in a few advantageous biological activities like anti-inflammatory,
anticoagulant and antitumor activities as well as low toxicity [103].
1422 R. S. Nandhini et al.
12.1 Coumarins
12.2 Flavonoids
and antineoplastic activities. Flavonoids can be extracted from high plants. They
can be found in yellow, orange, or red and so on is widely available as colourful
fruits and vegetables in human food: Apiaceae (parsley), Ericaceae (berries),
Rutaceae (orange fruit), Rosaceae (apple) and a famous sweet product of Theobroma
cacao, that is, black chocolate. Flavonoids have shown antineoplastic properties
work in several subjects.
12.3 Diarylheptanoid (Curcumin)
References
1. Wang ZB, Jiang H, Xia YG, Yang BY, Kuang HX (2012) α-Glucosidase inhibitory constitu-
ents from Acanthopanax senticosus harm leaves. Molecules 17(6):6269–6276
2. Chin WW, Parmentier J, Widzinski M, Tan EH, Gokhale R (2014) A brief literature and
patent review of nanosuspensions to a final drug product. J Pharm Sci 103(10):2980–2999.
https://doi.org/10.1002/jps.24098. Epub 2014 Aug 6. PMID: 25099918
3. Hwang KA, Park MA, Kang NH, Yi BR, Hyun SH, Jeung EB, Choi KC (2013) Anticancer
effect of genistein on BG-1 ovarian cancer growth induced by 17 β- estradiol or bisphenol A
via the suppression of the crosstalk between estrogen receptor alpha and insulin-like growth
factor-1 receptor signaling pathways. Toxicol Appl Pharmacol 272(3):637–646
4. Choi EJ, Jung JY, Kim GH (2014) Genistein inhibits the proliferation and differentiation of
MCF-7 and 3T3-L1 cells via the regulation of ERα expression and induction of apoptosis.
Exp Ther Med 8(2):454–458
5. Luo Y, Wang SX, Zhou ZQ, Wang Z, Zhang YG, Zhang Y, Zhao P (2014) Apoptotic effect
of genistein on human colon cancer cells via inhibiting the nuclear factor- kappa B (NF-κB)
pathway. Tumour Biol 35(11):11483–11488
6. Yamasaki M, Mine Y, Nishimura M, Fujita S, Sakakibara Y, Suiko M, Morishita K, Nishiyama
K (2013) Genistein induces apoptotic cell death associated with inhibition of the NF-κB
pathway in adult T-cell leukaemia cells. Cell Bio Int 37(7):742–747. https://doi.org/10.1002/
cbin.10101. Epub 2013 Apr 18. PMID: 23526666
7. Zhang Z, Wang CZ, Du GJ, Qi LW, Calway T, He TC et al (2013) Genistein induces G2/M
cell cycle arrest and apoptosis via ATM/p53-dependent pathway in human colon cancer cells.
Int J Oncol 43(1):289–296
8. Hirata H, Hinoda Y, Shahryari V, Deng G, Tanaka Y, Tabatabai ZL, Dahiya R (2014) Genistein
downregulates onco-miR-1260b and upregulates sFRP1 and Smad4 via demethylation and
histone modification in prostate cancer cells. Br J Cancer 110(6):1645–1654
9. Hirata H, Ueno K, Nakajima K, Tabatabai ZL, Hinoda Y, Ishii N, Dahiya R (2013) Genistein
downregulates onco-miR-1260b and inhibits Wnt-signalling in renal cancer cells. Br J Cancer
108(10):2070–2078
10. Chiyomaru T, Yamamura S, Fukuhara S, Hidaka H, Majid S, Saini S et al (2013) Genistein
up-regulates tumour suppressor microRNA-574-3p in prostate cancer. PLoS One 8(3):e58929
1424 R. S. Nandhini et al.
30. Reddy LH, Bazile D (2014) Drug delivery design for intravenous route with integrated physi-
cochemistry, pharmacokinetics and pharmacodynamics: illustration with the case of taxane
therapeutics. Adv Drug Deliv Rev 71:34–57
31. Ibrahim NK, Desai N, Legha S, Soon-Shiong P, Theriault RL, Rivera E et al (2002) Phase I
and pharmacokinetic study of ABI-007, a Cremophor-free, protein-stabilized, nanoparticle
formulation of paclitaxel. Clin Cancer Res 8(5):1038–1044
32. US Food and Drug Administration (2012) ABRAXANE® for injectable suspension (pacli-
taxel protein-bound particles for injectable suspension) (albumin-bound)
33. Roy U, Chakravarty G, Zu Bentrup KH, Mondal D (2009) Montelukast is a potent and dura-
ble inhibitor of multidrug resistance protein 2-mediated efflux of taxol and saquinavir. Biol
Pharm Bull 32(12):2002–2009
34. Javed B, Ikram M, Farooq F, Sultana T, Mashwani ZUR, Raja NI (2021) Biogenesis of sil-
ver nanoparticles to treat cancer, diabetes, and microbial infections: a mechanistic overview.
Appl Microbiol Biotechnol 105(6):2261–2275
35. Prakash NU, Bhuvaneswari S, Nandhini RS, Azeez NA, Al-Arfaj AA, Munusamy MA (2015)
Floral synthesis of silver nanoparticles using Stenolobium stans L. Asian J Chem 27(11):4089
36. Yesilot S, Aydin C (2019) Silver nanoparticles; a new hope in cancer therapy? East J Med
24(1):111–116
37. Alhadrami HA, Alkhatabi H, Abduljabbar FH, Abdelmohsen UR, Sayed AM (2021) Anticancer
potential of green synthesized silver nanoparticles of the soft coral c ladiellapachyclados sup-
ported by network pharmacology and in silico analyses. Pharmaceutics 13(11):1846
38. Hu XK, Rao SS, Tan YJ, Yin H, Luo MJ, Wang ZX et al (2020) Fructose-coated Angstrom
silver inhibits osteosarcoma growth and metastasis via promoting ROS-dependent apoptosis
through the alteration of glucose metabolism by inhibiting PDK. Theranostics 10(17):7710
39. Mengesha AE, Youan BC (2013) Nanodiamonds for drug delivery systems. In: Diamond-
based materials for biomedical applications. Woodhead Publishing, Elsevier, Sawston,
Cambridge, UK, pages 186–205
40. Mochalin VN, Shenderova O, Ho D, Gogotsi Y (2012) The properties and applications of
nanodiamonds. Nat Nanotechnol 7(1):11–23
41. Horie M, Komaba LK, Kato H, Nakamura A, Yamamoto K, Endoh S et al (2012) Evaluation
of cellular influences induced by stable nanodiamond dispersion; the cellular influences of
nanodiamond are small. Diam Relat Mater 24:15–24
42. Gwak R, Lee GJ, Kim H, Lee MK, Rhee CK, Dae-Ro C et al (2015) Efficient doxorubicin
delivery using deaggregated and carboxylatednanodiamonds for cancer cell therapy. Nanosci
Nanotechnol Lett 7(9):723–728
43. Xiao J, Duan X, Yin Q, Zhang Z, Yu H, Li Y (2013) Nanodiamonds-mediated doxorubicin
nuclear delivery to inhibit lung metastasis of breast cancer. Biomaterials 34(37):9648–9656
44. National Center for Biotechnology Information (2022) PubChem compound summary for
CID 2775, Citropten. Retrieved February 25, 2022 from https://pubchem.ncbi.nlm.nih.gov/
compound/Citropten
45. Hofheinz RD, Gnad-Vogt SU, Beyer U, Hochhaus A (2005) Liposomal encapsulated anti-
cancer drugs. Anti-Cancer Drugs 16(7):691–707
46. Silverman JA, Deitcher SR (2013) Marqibo(vincristine sulfate liposome injection) improves
the pharmacokinetics and pharmacodynamics of vincristine. Cancer Chemother Pharmacol
71(3):555–564
47. Castle MC, Mead JAR (1978) Investigations of the metabolic fate of tritiated vincristine in
the rat by high-pressure liquid chromatography. Biochem Pharmacol 27(1):37–44
48. Krishna R, Webb MS, Onge GS, Mayer LD (2001) Liposomal and nonliposomal drug phar-
macokinetics after administration of liposome-encapsulated vincristine and their contribution
to drug tissue distribution properties. J Pharmacol Exp Ther 298(3):1206–1212
49. Webb MS, Harasym TO, Masin D, Bally MB, Mayer LD (1995) Sphingomyelin-cholesterol
liposomes significantly enhance the pharmacokinetic and therapeutic properties of vincris-
tine in murine and human tumour models. Br J Cancer 72(4):896–904
1426 R. S. Nandhini et al.
50. Hagemeister F, Rodriguez MA, Deitcher SR, Younes A, Fayad L, Goy A, Cabanillas F (2013)
Long term results of a phase 2 study of vincristine sulfate liposome injection (M arqibo)
substituted for non-liposomal vincristine in cyclophosphamide, doxorubicin, vincristine,
prednisone with or without rituximab for patients with untreated aggressive non-H odgkin
lymphomas. Br J Haematol 162(5):631–638
51. Ye L, He J, Hu Z, Dong Q, Wang H, Fu F, Tian J (2013) Antitumor effect and toxicity of
Lipusu in rat ovarian cancer xenografts. Food Chem Toxicol 52:200–206
52. Xu X, Wang L, Xu HQ, Huang XE, Qian YD, Xiang J (2013) Clinical comparison between
paclitaxel liposome (Lipusu) and paclitaxel for treatment of patients with metastatic gastric
cancer. Asian Pac J Cancer Prev 14(4):2591–2594
53. Cabral H, Kataoka K (2014) Progress of drug-loaded polymeric micelles into clinical studies.
J Control Release 190:465–476
54. Kim SC, Kim DW, Shim YH, Bang JS, Oh HS, Kim SW, Seo MH (2001) In vivo evalua-
tion of polymeric micellar paclitaxel formulation: toxicity and efficacy. J Control Release
72(1–3):191–202
55. Lee KS, Chung HC, Im SA, Park YH, Kim CS, Kim SB et al (2008) Multicenter phase II trial
of Genexol-PM, a Cremophor-free, polymeric micelle formulation of paclitaxel, in patients
with metastatic breast cancer. Breast Cancer Res Treat 108(2):241–250
56. Kim TY, Kim DW, Chung JY, Shin SG, Kim SC, Heo DS, Kim NK, Bang YJ (2004) Phase I
and pharmacokinetic study of Genexol-PM, a cremophor-free, polymeric micelle-formulated
paclitaxel, in patients with advanced malignancies. Clin Cancer Res 10(11):3708–3716.
https://doi.org/10.1158/1078-0432.CCR-03-0655. PMID: 15173077
57. DeLano WL (2002) Pymol: an open-source molecular graphics tool. CCP4 Newsl Protein
Crystallogr 40(1):82–92
58. Semwal DK, Semwal RB, Combrinck S, Viljoen A (2016) Myricetin: a dietary molecule with
diverse biological activities. Nutrients 8(2):90
59. Mohan UP, Sriram B, Panneerselvam T, Devaraj S, Mubarak Ali D, Parasuraman P et al
(2020) Utilization of plant-derived Myricetin molecule coupled with ultrasound for the syn-
thesis of gold nanoparticles against breast cancer. Naunyn Schmiedeberg’s Arch Pharmacol
393(10):1963–1976
60. Fern K (2018) Tropical plants database, Ken Fern. tropical. theferns. info
61. Gu FX, Karnik R, Wang AZ, Alexis F, Levy-Nissenbaum E (2007) Targeted nanoparticles for
cancer treatment. Nano Today 2:14–21
62. Cho K, Wang X, Nie S, Chen Z, Shin DM (2008) Therapeutic nanoparticles. Clin Cancer Res
14(5):1310–1316
63. Mishra B, Patel BB, Tiwari S (2009) Colloidal nanocarriers: a review on formulation technol-
ogy, types and applications toward targeted drug delivery. Nanomedicine: NBM 1:17
64. Lim KJ, Bisht S, Bar EE, Maitra A, Eberhart CG (2011) A polymeric nanoparticle formula-
tion of curcumin inhibits growth, clonogenicity and stem-like fraction in malignant brain
tumors. Cancer Biol Ther 11(5):464–473
65. Zu Y, Wang D, Zhao X, Jiang R, Zhang Q, Zhao D et al (2011) A novel preparation method
for camptothecin (CPT) loaded folic acid conjugated dextran tumor- targeted nanoparticles.
Int J Mol Sci 12(7):4237–4249
66. Pimple S, Manjappa AS, Ukawala M, Murthy RSR (2012) PLGA nanoparticles loaded with
etoposide and quercetin dihydrate individually: in vitro cell line study to ensure advantage of
combination therapy. Cancer Nanotechnol 3(1):25–36
67. Siu YS, Li L, Leung MF, Lee KLD, Li P (2012) Polyethylenimine-based amphiphilic core–
shell nanoparticles: study of gene delivery and intracellular trafficking. Biointerphases 7(1):16
68. Tang X, Cai S, Zhang R, Liu P, Chen H, Zheng Y, Sun L (2013) Paclitaxel- loaded nanopar-
ticles of star-shaped cholic acid-core PLA-TPGS copolymer for breast cancer treatment.
Nanoscale Res Lett 8(1):1–12
69. Sundar VD, Dhanaraju MD, Sathyamoorthy N (2014) Fabrication and characterization of
etoposide loaded magnetic polymeric microparticles. Int J Drug Deliv 6(1):24
Use of Plant-Derived Nanoparticles in Cancer Therapy 1427
70. Han FY, Thurecht KJ, Whittaker AK, Smith MT (2016) Bioerodable PLGA- based micropar-
ticles for producing sustained-release drug formulations and strategies for improving drug
loading. Front Pharmacol 7:185
71. Yang A, Liu Z, Yan B, Zhou M, Xiong X (2016) Preparation of camptothecin- loaded target-
ing nanoparticles and their antitumor effects on hepatocellular carcinoma cell line H22. Drug
Deliv 23(5):1699–1706
72. Zhou H, Liu X, Wu F, Zhang J, Wu Z, Yin H, Shi H (2016) Preparation, characterization,
and antitumor evaluation of electrospun resveratrol loaded nanofibers. J Nanomaterials 2016
73. Wong HL, Bendayan R, Rauth AM, Li Y, Wu XY (2007) Chemotherapy with anticancer
drugs encapsulated in solid lipid nanoparticles. Adv Drug Deliv Rev 59(6):491–504
74. Ekambaram P, Sathali AAH, Priyanka K (2012) Solid lipid nanoparticles: a review. Sci Rev
Chem Commun 2(1):80–102
75. Yassin AEB, Albekairy A, Alkatheri A, Sharma RK (2013) Anticancer- loaded solid lipid
nanoparticles: high potential advancement in chemotherapy. Digest J Nanomater Biostruct
(DJNB) 8(2):905–916
76. Abd-Allah FI, Dawaba HM, Samy AM, Nutan MT (2014) Application of solvent injection
method to develop stable, sustained release solid lipid nanoparticles of curcumin. Int J Dev
Res 4:2734–2742
77. Chadha R, Kapoor VK, Thakur D, Kaur R, Arora P, Jain DVS (2008) Drug carrier systems for
anticancer agents: a review. J Sci Ind Res 67:185–197
78. Narayanan NK, Nargi D, Randolph C, Narayanan BA (2009) Liposome encapsulation of cur-
cumin and resveratrol in combination reduces prostate cancer incidence in PTEN knockout
mice. Int J Cancer 125(1):1–8
79. Ramana LN, Sharma S, Sethuraman S, Ranga U, Krishnan UM (2012) Investigation on the
stability of saquinavir loaded liposomes: implication on stealth, release characteristics and
cytotoxicity. Int J Pharm 431(1–2):120–129
80. Venegas B, Zhu W, Haloupek NB, Lee J, Zellhart E, Sugár IP, Kiani MF, Chong PL (2012)
Cholesterol superlattice modulates CA4P release from liposomes and CA4P cytotoxic-
ity on mammary cancer cells. Biophys J 102(9):2086–2094. https://doi.org/10.1016/j.
bpj.2012.03.063. PMID: 22824272; PMCID: PMC3341537
81. Shah SM, Goel PN, Jain AS, Pathak PO, Padhye SG, Govindarajan S, Ghosh SS, Chaudhari
PR, Gude RP, Gopal V, Nagarsenker MS (2014) Liposomes for targeting hepatocellular carci-
noma: use of conjugated arabinogalactan as targeting ligand. Int J Pharm 477(1–2):128–139.
https://doi.org/10.1016/j.ijpharm.2014.10.014. Epub 2014 Oct 11. PMID: 25311181
82. Mehrabi M, Esmaeilpour P, Akbarzadeh A, Saffari Z, Farahnak M, Farhangi A, Chiani M
(2016) Efficacy of pegylated liposomal etoposide nanoparticles on breast cancer cell lines.
Turk J Med Sci 46(2):567–571
83. Li R, Wu RA, Zhao L, Hu Z, Guo S, Pan X, Zou H (2011) Folate and iron difunctional-
ized multiwall carbon nanotubes as dual-targeted drug nanocarrier to cancer cells. Carbon
49(5):1797–1805
84. Tian Z, Shi Y, Yin M, Shen H, Jia N (2011) Functionalized multiwalled carbon nanotubes-
anticancer drug carriers: synthesis, targeting ability and antitumor activity. Nano Biomed
Eng 3(3)
85. Popov VN (2004) Carbon nanotubes: properties and application. Mater Sci Eng R: Rep
43(3):61–102
86. Nakanishi T, Fukushima S, Okamoto K, Suzuki M, Matsumura Y, Yokoyama M et al (2001)
Development of the polymer micelle carrier system for doxorubicin. J Control Release
74(1–3):295–302
87. Husseini GA, Pitt WG (2008) Micelles and nanoparticles for ultrasonic drug and gene deliv-
ery. Adv Drug Deliv Rev 60(10):1137–1152
88. Maeda H, Bharate GY, Daruwalla J (2009) Polymeric drugs for efficient tumor targeted drug
delivery based on EPR effect. Eur J Pharm Biopharm 71(3):409
1428 R. S. Nandhini et al.
89. Mourya VK, Inamdar N, Nawale RB, Kulthe SS (2011) Polymeric micelles: general consid-
erations and their applications. Indian J Pharm Educ Res 45(2):128–138
90. Wang C, Feng L, Yang X, Wang F, Lu W (2013) Folic acid-conjugated liposomal vincristine
for multidrug resistant cancer therapy. Asian J Pharm Sci 8(2):118–127
91. Kore G, Kolate A, Nej A, Misra A (2014) Polymeric micelle as multifunctional pharmaceuti-
cal carriers. J Nanosci Nanotechnol 14(1):288–307. https://doi.org/10.1166/jnn.2014.9021
92. Naha PC, Davoren M, Lyng FM, Byrne HJ (2010) Reactive oxygen species (ROS) induced
cytokine production and cytotoxicity of PAMAM dendrimers in J774A. 1 cells. Toxicol Appl
Pharmacol 246(1–2):91–99
93. Abdel-Rahman MA, Al-Abd AM (2013) Thermoresponsive dendrimers based on oligoethyl-
ene glycols: design, synthesis and cytotoxic activity against MCF-7 breast cancer cells. Eur
J Med Chem 69:848–854
94. Baig T, Nayak J, Dwivedi V, Singh A, Srivastava A, Tripathi PK (2015) A review about den-
drimers: synthesis, types, characterization and applications. Int J Adv Pharm Biol Chem
4(1):44–59
95. Malar CG (2015) Dendrosomal capsaicin nanoformulation for the invitro anticancer effect on
HEp 2 and MCF-7 cell lines. Int J Appl Bioeng 9(2)
96. Yang Q, Yang Y, Li L, Sun W, Zhu X, Huang Y (2015) Polymeric nanomedicine for tumor-
targeted combination therapy to elicit synergistic genotoxicity against prostate cancer. ACS
Appl Mater Interfaces 7(12):6661–6673
97. Kuppusamy P, Yusoff MM, Maniam GP, Govindan N (2016) Biosynthesis of metallic
nanoparticles using plant derivatives and their new avenues in pharmacological applications–
an updated report. Saudi Pharma J 24(4):473–484
98. Melo ISVD, Santos AFD, Lemos TLGD, Goulart MOF, Santana AEG (2015) Oncocalyxone
A functions as an anti-glycation agent in vitro. PLoS One 10(6):e0131222
99. Barreto AC, Santiago VR, Freire RM, Mazzetto SE, Denardin JC, Mele G et al (2013)
Magnetic nanosystem for cancer therapy using oncocalyxone a, an antitomour secondary
metabolite isolated from a Brazilian plant. Int J Mol Sci 14(9):18269–18283
100. Cavalcanti IDL, Ximenes RM, Pessoa ODL, Magalhães NSS, de Britto Lira-Nogueira MC
(2021) Fucoidan-coated PIBCA nanoparticles containing oncocalyxone A: activity against
metastatic breast cancer cells. J Drug Deliv Sci Technol 65:102698
101. Pessoa C, Vieira FMAC, Lemos TG, Moraes MO, Lima PDL, Rabenhorst SHB et al (2003)
Oncocalyxone A from Auxemmaoncocalyx lacks genotoxic activity in phytohemagglutinin-
stimulated lymphocytes. Teratog Carcinog Mutagen 23(S1):215–220
102. Sbardelotto AB (2013) Estudo do mecanismo de citotóxicidade da oncocalixona-A emleuce-
miapromiolocíticahumana–linhagem HL-60
103. Lira MCB, Santos-Magalhães NS, Nicolas V, Marsaud V, Silva MPC, Ponchel G, Vauthier C
(2011) Cytotoxicity and cellular uptake of newly synthesized fucoidan-coated nanoparticles.
Eur J Pharm Biopharm 79(1):162–170
104. Zhang Z, Teruya K, Yoshida T, Eto H, Shirahata S (2013) Fucoidan extract enhances the anti-
cancer activity of chemotherapeutic agents in MDA-MB-231 and MCF-7 breast cancer cells.
Mar Drugs 11(1):81–98
The Global Concern for Cancer
Emergence and Its Prevention:
A Systematic Unveiling of the Present
Scenario
1 Introduction
Cancer is a major public health issue and the leading cause of death globally,
accounting for about 10.0 million cancer deaths worldwide in 2020 [1–3]. It remains
the second leading cause of morbidity and mortality after cardiovascular disease
around the globe, and predominantly all cancer cases have their roots in lifestyle
and environment. The lifestyle factors comprising diet, obesity, cigarette smoking,
alcohol consumption, stress, physical inactivity and continuous exposure of human
beings to certain infections type, carcinogenic agents, ionizing radiations, and envi-
ronmental contaminants or toxicants [4–6]. Even though the world is paying good
attention to control the spread of infectious diseases, the growing burden of cancer
is being ignored in developing and least developed countries, where survival rates
are often less than half and there is also a lack of willingness and preparation among
the governing authorities to combat cancer [7, 8]. Over 100 different forms of can-
cer exist that influence all communities worldwide; however, there are marked vari-
ations in the types and pervasiveness of cancers among groups. Scientific data
suggests that new cancer cases are constantly being reported and a 47% rise from
2020 is expected in the global cancer burden, which is likely to be 28.4 million
cases by 2040 [9]. Although the total burden of cancer remains highest in rich
income civilizations, less developed and developing economies are reducing the gap
very quickly. As developing nations succeed in attaining lifestyles similar to those
M. S. Ghufran (*)
Department of Chemistry, Faculty of Natural and Computational Sciences, Gambella
University, Gambella, Federal Democratic Republic of Ethiopia
e-mail: [email protected]
P. Soni · G. R. Duddukuri
Department of Biochemistry and Molecular Biology, School of Biological Sciences, Central
University of Kerala, Tejaswini Hills, Kasargod, Kerala, India
of advanced nations, the risk of encountering cancer rates is getting much higher,
specifically breast, uterus, colon, and prostate cancer [10]. About one-third of deaths
from cancer are caused by obesity, high body mass index, low intake of fruits and
vegetables, use of tobacco, excessive alcohol consumption, and lack of physical
activity [11]. Certain types of infections that causes cancer such as human papillo-
mavirus (HPV) and hepatitis are accountable for 30% of the total cancer cases in
low- and lower-middle-income countries (LMICs) [12]. Cancer has a great impact
on the country’s economy and continues to grow. For example, in 2010, the total
annual financial cost of cancer was estimated at 1.16 trillion US dollars [13]. The
increasing prevalence of cancer as a foremost reason of death partially discloses the
significant decline in mortality rates of coronary heart disease and stroke in com-
parison to cancer in various countries [14]. Overall, the incidence of cancer and
mortality is growing fast around the globe, which reveals the growth and aging of
the population together as well as changes in the frequency and distribution of the
primary risk factors. Many of them are associated with social and economic devel-
opment [15, 16]. In this chapter, we summarized the available data to explain the
severity of the global cancer burden. We have accessed the GLOBOCAN 2020 data-
base created by the International Agency for Research on Cancer (IARC) to evalu-
ate the morbidity and mortality of cancer in different geographical regions of
the world.
over 47% global cancer burden estimated for the next 20 years [2, 20]. Breast cancer
has overtaken lung cancer as the most commonly recognized cancer in women.
Global comprehensive data on multiple forms of cancer in 2020 is shown in Table 1.
The top five most common new cases of cancer reported in 2020 across the world
Table 1 Estimated worldwide number of cases in terms of incidence, mortality, and prevalence by
cancer site in both men and women in 2020
Estimated new cases in Estimated deaths in 5-year prevalent cases
Types of cancer 2020 2020 (all ages)
Breast 2,261,419 684,996 7,790,717
Lung 2,206,771 1,796,144 2,604,791
Prostate 1,414,259 375,304 4,956,901
Colorectum 1,880,725 915,880 5,111,957
Stomach 1,089,103 768,793 1,805,968
Liver 905,677 830,180 994,539
Cervix uteri 604,127 341,831 1,495,211
Esophagus 604,100 544,076 668,388
Thyroid 586,202 43,646 1,984,927
Bladder 573,278 212,536 1,720,625
Non-Hodgkin 544,352 259,793 1,544,488
lymphoma
Pancreas 495,773 466,003 379,958
Leukemia 474,519 311,594 1,340,506
Kidney 431,288 179,368 1,207,547
Corpus uteri 417,367 97,370 1,415,213
Lip, oral cavity 377,713 177,757 959,248
Melanoma (skin) 324,635 57,043 1,092,818
Ovary 313,959 207,252 823,315
Brain, CNS 308,102 251,329 837,152
Larynx 184,615 99,840 518,380
Multiple myeloma 176,404 117,077 450,579
Nasopharynx 133,354 80,008 382,507
Gallbladder 115,949 84,695 137,466
Oropharynx 98,412 48,143 258,543
Hypopharynx 84,254 38,599 132,717
Hodgkin lymphoma 83,087 23,376 281,112
Testis 74,458 9334 296,686
Salivary glands 53,583 22,778 160,292
Anus 50,865 19,293 141,378
Vulva 45,240 17,427 135,892
Penis 36,068 13,211 102,157
Kaposi sarcoma 34,270 15,086 82,033
Mesothelioma 30,870 26,278 37,047
Vagina 17,908 7995 44,613
Data source: GLOBOCAN 2020
Abbreviation: CNS central nervous system
1432 M. S. Ghufran et al.
were breast (2.26 million cases), lung (2.21 million), prostate (1.41 million), colon
(1.15 million), and cancer of the stomach (1.09 million). Figure 1 shows the global
distribution of the top 10 cancer types for the estimated number of cancer incidence
and mortality in men and women in 2020. The most common reason for deaths from
cancer in 2020 were reported for the lungs (1.80 million), followed by colon and
rectum (0.93 million), liver (0.83 million), stomach (0.77 million), and breast (0.68
million) [9, 22]. The data summarized in Table 2 provides the status and trends in
organ-based cancer incidence and mortality cases in India for the year 2020. The top
five new cancer cases reported were cancer of the female breast, lip, oral cavity,
Fig. 1 Distribution of cancer incidence and mortality in the world for the top 10 most common
cancers in 2020. (a) Both sexes, (b) females, and (c) males. The area of the pie diagram represents
the proportion of the total number of cancer cases or deaths. (Data Source: GLOBOCAN 2020.
https://gco.iarc.fr/today/home)
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1433
Table 2 Estimated number of cases in India in terms of incidence, mortality, and prevalence,
based on site of cancer, in both men and women in the year 2020
Estimated new cases in Estimated deaths in 5-year prevalent cases
Types of cancer 2020 2020 (all ages)
Breast 178,361 90,408 459,271
Lip oral cavity 135,929 75,290 300,413
Cervix uteri 123,907 77,348 283,842
Lung 72,510 66,279 80,817
Esophagus 63,180 58,342 68,607
Stomach 60,222 53,253 81,270
Leukemia 48,419 35,392 127,493
Ovary 45,701 32,077 103,716
Non-Hodgkin 35,828 20,390 88,272
lymphoma
Liver 34,743 33,793 38,602
Larynx 34,687 21,660 82,087
Prostate 34,540 16,783 67,909
Colorectum 59,906 35,385 128,320
Brain, CNS 31,460 26,656 74,398
Hypopharynx 28,489 11,443 39,750
Bladder 21,096 11,154 49,257
Oropharynx 20,617 12,703 44,398
Thyroid 20,432 4895 55,248
Gallbladder 19,570 14,736 25,138
Kidney 16,861 9897 39,150
Corpus uteri 16,413 6385 43,484
Multiple myeloma 14,641 12,556 30,640
Pancreas 12,642 12,153 11,928
Penis 10,677 4760 26,280
Hodgkin lymphoma 9221 3513 24,928
Salivary glands 7850 5127 20,448
Nasopharynx 5697 4148 14,196
Vagina 5518 2723 12,315
Anus 5452 2776 12,278
Testis 4681 1252 14,812
Melanoma (skin) 3916 2296 9637
Vulva 3447 1694 8928
Mesothelioma 1709 1543 2223
Kaposi sarcoma 66 43 156
Data source: GLOBOCAN 2020
Abbreviation: CNS central nervous system
cervix uteri, lung, and esophagus. Figure 2 depicts the percentage distribution of the
top 10 cancer types in 2020 for the estimated number of cancer incidence and mor-
tality rates in men and women in India.
Reports suggests that in 2018, approximately 18.1 million people across the
world were suffering from cancer, and 9.6 million people died from the disease. By
1434 M. S. Ghufran et al.
Fig. 2 Distribution of cancer incidence and mortality in India for the top 10 most common cancers
in 2020. (a) Both sexes, (b) females, and (c) males. The area of the pie diagram represents the
proportion of the total number of cancer cases or deaths. (Data Source: GLOBOCAN 2020. https://
gco.iarc.fr/today/home)
the year 2040, these numbers will become twofold to nearby 29–37 million new
cases of cancer, with the highest upsurge and incidence of over two-thirds of the
world’s cancer in LMICs [20]. Among adults aged between 30 and 69 years, cancer
is the main reason for roughly 30% of all premature deaths from non-communicable
diseases (NCDs). In the year 2018, lung cancer (11.6% of total cases) was most
commonly detected cancer followed by female breast and colorectal cancer with
11.6% and 10.2% of all reported cases, respectively. The leading cause of death was
lung cancer (18.4% of total deaths) followed by colorectal and stomach cancers
with reported deaths of 9.2% and 8.2%, respectively [20]. The 10 most common
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1435
cancers namely lung, breast, colorectum, prostate, stomach, liver, esophagus, cervix
uteri, thyroid, and bladder were accountable for 60–70% of cancer incidence and
death [20]. The data provided in Table 3 shows incidence and mortality in the year
2020 based on age standardized rates and cumulative risk for various cancer cases
in different regions of the world.
Substantial global diversity among major cancer types is because of variances in life
expectancy and exposure to risk factors [18]. For the majority of cancer types, the
age-standardized rates (ASR) are higher in countries with a higher human develop-
ment index (HDI); however, in countries with low HDI some cancers namely naso-
pharyngeal cancer, cervical cancer, and Kaposi sarcoma are more common [2, 23].
Recent trends in the rates of major cancer acquired from high-quality cancer regis-
try data in nations based on the Human Development Index suggest that increasing
age-standardized rates contribute to two-fifths and aging of the population with
increased life expectancy contributes three-fifths of the future burden of cancer [24].
In absolute terminologies, the cancer burden is highest in countries with high and
very high Human Development Indexes (HDIs); however, the rises will be propor-
tionally more in terms of relative aspect in countries with low and medium HDIs
[3]. Particularly, in nations with low or medium HDI, the expected rise in the annual
number of new cancer patients in the coming decades is a clear sign of immediate
and good investment in control of cancer. Cancer cases in children are less com-
monly recognized, resulting in significant global dissimilarity. Worldwide, more
than 0.3 million children are diagnosed with cancer each year, and the frequency is
likely to rise with control of competing reasons of death, such as communicable
diseases [25].
There has been an epidemiological transition in the past 60 years, as the mortality
from contagious diseases has declined and the burden of non-communicable dis-
eases, including cancer, has augmented [20]. This epidemiological shift is due to the
discovery of antibiotics, improvements in sanitation and the development of vac-
cines. The trends in cancer type with implications of strategies to control cancer are
strongly affected by changes in prevalence and the risk factor distribution. For
instance, the various effects of the smoking epidemic on the occurrence of lung
cancer are associated with the prevalence of tobacco usage in nations with high and
lower HDIs [26]. Likewise, the incidence of cancers associated with the infection,
1436 M. S. Ghufran et al.
Table 3 Global incidence and mortality rates (age-standardized rate, cumulative risk) for different
cancer cases in both sexes in the year 2020
Incidence Mortality
Both sexes Both sexes
Cumulative Cumulative
Age- risk ages Age- risk ages
World standardized 0–74 years standardized 0–74 years
region New cases rate (World) (%) Deaths rate (world) (%)
Eastern 331,233 130.2 13.6 222,189 92.1 9.91
Africa
Middle 106,467 111.1 11.7 71,570 78.4 8.4
Africa
Northern 307,507 141.9 14.61 191,081 89.9 9.21
Africa
Southern 116,391 202.4 20.0 61,659 109.0 11.5
Africa
Western 247,611 111.5 11.7 164,930 78.8 8.44
Africa
Caribbean 113,280 191.7 19.72 65,954 102.7 10.44
Central 261,646 140.2 14.3 126,071 66.0 6.91
America
South 1,095,348 201.4 20.24 521,389 91.5 9.5
America
Northern 2,556,862 360.7 33.93 699,274 87.1 9.22
America
Eastern 6,008,355 217.2 21.91 3,617,104 123.2 13.1
Asia
China 4,568,754 204.8 21.0 3,002,899 129.4 13.9
South- 1,100,037 152.1 15.62 689,093 95.4 10.1
Eastern
Asia
South- 1,951,843 102.5 10.93 1,258,683 67.0 7.41
Central
Asia
India 1,324,413 97.5 10.45 851,678 63.2 7.1
Western 443,475 175.8 18.31 244,551 98.6 10.6
Asia
Central and 1,314,193 246.1 25.5 695,828 118.9 13.4
Eastern
Europe
Western 1,424,394 325.0 31.24 559,671 103.3 10.85
Europe
Southern 953,048 279.2 27.4 423,090 98.9 10.52
Europe
Northern 706,808 316.5 30.5 276,642 99.8 10.3
Europe
(continued)
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1437
Table 3 (continued)
Incidence Mortality
Both sexes Both sexes
Cumulative Cumulative
Age- risk ages Age- risk ages
World standardized 0–74 years standardized 0–74 years
region New cases rate (World) (%) Deaths rate (world) (%)
Australia/ 235,955 447.6 40.43 58,744 85.8 8.55
New
Zealand
Melanesia 15,658 195.9 20.0 9022 120.7 12.6
Polynesia 1668 230.0 24.0 927 127.0 13.93
Micronesia 1010 180.6 18.85 661 118.1 12.7
Low HDI 650,423 115.7 12.1 439,852 82.7 8.8
Medium 2,326,749 108.5 11.52 1,513,219 71.5 7.9
HDI
High HDI 7,371,321 190.5 19.53 4,521,833 113.7 12.23
Very high 8,934,818 295.3 28.93 3,478,767 98.7 10.4
HDI
World 19,292,789 201.0 20.44 9,958,133 100.7 10.7
Low 447,703 121.2 12.5 308,783 87.5 9.2
income
Low middle 3,099,791 115.2 12.3 1,982,660 74.9 8.2
income
Upper 7,894,392 197.2 20.2 4,771,627 115.2 12.4
middle
income
High 7,841,425 309.7 30.1 2,890,601 94.8 9.9
income
Data source: GLOBOCAN 2020
Abbreviations: CNS central nervous system, HDI human development index
for example liver, cervix, and cancer of the stomach, has reduced significantly in the
last three decades in nations with very high HDIs, but they are common in countries
with low and medium HDIs [20, 27]. Trends in death rates are also a function of
changes in survival and occurrence that are linked to the ability of the health system
to manage cancer, including previous screening and access to effective treatment.
There are two clear findings from the studies on the chances of survival of cancer:
(i) investing in cancer management can improve survival and (ii) survival trends
vary by country and region [27].
1438 M. S. Ghufran et al.
Health determinants such as economic status, biological factors, physical and social
environments have a significant impact on cancer incidence and its outcomes. For
instance, in countries with higher HDIs, use of tobacco is more prevalent in groups
with low socioeconomic status, ensuing in 60–90% higher degrees of cancers asso-
ciated with tobacco [12, 28]. Environmental factors also play a major role in the
socioeconomic disparity of cancer. The prevalence of many common cancers asso-
ciated with infection such as cancers of the liver (hepatitis B and C viruses), cervix
(Human papillomavirus), and stomach (Helicobacter pylori), is higher in the most
underprivileged groups both at the national and sub-national level with larger over-
all proportion of cancers associated with infection in lower socio-economic groups
and low-income nations [12, 28]. People living with HIV also have a higher risk of
developing many types of cancer, specifically those associated with AIDS (invasive
cervical cancer, non-Hodgkin lymphomas and Kaposi sarcoma) and transmissible
agents (liver cancer associated to hepatitis B and C viruses, Hodgkin lymphoma
linked to Epstein-Barr viruses and anal cancer caused by Human papillomavirus)
[29, 30]. By means of better treatment and a projected rise in people’s life expec-
tancy living with HIV/AIDS, more investment in clinical trials will be needed to
detect the best effective control strategies for cancer prevention.
Death from cancer is closely associated with the cancer stage at the time of diag-
nosis and the quality of treatment being given. The deprived groups participate less
in the screening programs and often experience geographical and economic barriers
which result in very late diagnoses and significant variances in outcomes [31–33].
Even if cancer is detected at an initial stage, these groups are more likely to get poor
quality care or fail to complete treatment or, in case of poor prognosis of cancers, be
transitioned to non-palliative care [34–36].
Cancer is a disease where normal cells are transformed into abnormal tumor cells in
a multistage process that typically develops from a precancerous lesion into a malig-
nant tumor. These alterations are the consequence of the interaction between genetic
factors of the individuals and different classes of external agents or environmental
factors or cancer-causing agents (Fig. 3). The contribution of environmental and
genetic factors toward the risk of cancer emergence is 90–95% and 5–10%, respec-
tively. These risk factors as mentioned by the WHO [22, 37] includes:
• Physical carcinogenic agents, such as exposure to electromagnetic fields, ioniz-
ing and ultraviolet radiation.
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1439
Fig. 3 Schematic representation showing the influence of environmental and genetic factors in the
emergence of cancer. Cancer is a disorder of altered cell differentiation and growth, arises due to
the consequence of the interaction between genetic factors of individuals and different classes of
environmental factors. The contribution of environmental and genetic factors towards the risk of
cancer emergence is 90–95% and 5–10%, respectively
3.1 Physical Factors
3.1.1 Ionizing Radiation
Ionizing radiation is one of the commonly listed potential carcinogens and its low
dose has enough energy to damage the DNA and may induce tumors in all organs of
the body. Individuals are more likely to get an increased dose of radiation exposure
during diagnostic imaging, medical treatment and therapeutic reasons or during
educational and scientific research in academia, which ultimately leads to the risk of
developing cancer [38, 39]. Most solid cancers and leukemia are associated with
radiation exposure which depends upon the dose and number of exposures. In gen-
eral, the risk of developing cancer as a result of a single low-dose exposure of 0.1
gray (Gy) was estimated to be approximately 1%. The estimates of individuals
1440 M. S. Ghufran et al.
exposed at a young age are large; for instance, the lifetime risk of individuals
exposed at age 10 would be about 2% [38]. Studies have found that the percentage
of all cancers caused by radiation is 2–3%, and children exposed to X-rays often
develop leukemia, liver, and bone cancer [40, 41]. Ionizing radiation can also stimu-
late neoplasia in the case of penetrating irradiation of the bladder, chest, and thyroid
gland. Radiation therapy in childhood stimulates tumorigenesis in the digestive tract
and the risk of developing malignant gastrointestinal tumors [41].
3.1.3 Ultraviolet Radiation
Another most common environmental factor affecting the skin is ultraviolet (UV)
radiation, which is primarily responsible for its damaging nature and is considered
as a potential risk factor causing melanoma and other skin malignancies [48].
Chronic and overexposure to UV rays from the sun can often cause adverse early-
onset symptoms, especially sunburn and late signs of accelerated skin aging. Most
skin cancers are caused by exposure to UV radiations in the sunlight. Epidemiological
and molecular studies have validated that extreme exposure to UV radiation is asso-
ciated with the increased risk of three most common types of skin cancers such as
malignant melanoma, squamous skin carcinoma, and basal cell carcinoma [49]. UV
radiation induces the formation of thymine-thymine pyrimidine-pyrimidone (6-4)
photoproduct (T (6-4) T) and thymine-thymine cyclobutane dimer (T-T), thereby
causing damage to the DNA of epidermal cells [50, 51]. These DNA adducts are
mutagenic, if left unrepaired, get incorporated into the DNA and eventually inter-
fere with the apoptotic pathways and induce cellular proliferation leading to tumor
development [50–52].
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1441
3.2 Chemical Factors
3.2.1 Alcohol Consumption
3.2.2 Tobacco Use
Tobacco smoking is a significant risk factor and the principal cause of lung carcino-
genesis, which causes more than seven million deaths every year worldwide.
Tobacco smoking accounts for approximately 80–90% of lung cancer in the United
States. It also causes several other types of cancer such as the colon and rectum,
cervix, pancreas, stomach, liver, kidney, bladder, throat, esophagus, mouth, and lar-
ynx [58]. All tobacco products, partly or completely made of tobacco leaves, are
sources of many carcinogens and other toxic substances that damage DNA. The
recent comprehensive statistical data on global trends in cigarette smoking high-
lights that the number of smokers continues to rise and has increased to 1.1 billion
in 2019, among which approximately 90% are new smokers who became addicted
by the age of 25 years [59]. Globally, tobacco use resulted in approximately 7.7 mil-
lion deaths in 2019 out of which nearly 1.3 million people lost their lives due to
bronchial, tracheal, and lung cancer [60]. The findings of the report also suggest that
1442 M. S. Ghufran et al.
if the smoking pattern does not change across the world, the condition would be
more horrible as more than eight million people will die every year from diseases
associated with tobacco use by 2030 [61]. Smokeless tobacco products are a major
threat and create a global burden of disease. Smokeless tobacco refers to various
products containing tobacco consumed by sniffing, chewing, or keeping it in the
mouth. It is being used across the world and poses a major public health issue in
South and South-East Asia as the highest rate of consumption was noticed in these
regions. In 2017, approximately 0.098 million people worldwide died because of
esophageal, pharyngeal, and oral cancers [62]. According to a new report published
by the National Cancer Registry of India (NCR), which works under the Indian
Council of Medical Research (ICMR), cancer cases attributable to tobacco con-
sumption were responsible for more than a quarter, or about 27% of India’s cancer
burden in 2020 [63].
Human beings often come into contact with many chemicals used in domestic life,
the environment, and agriculture. The discharge of toxic wastes or pollutants from
industries and the emissions of automobile exhaust contain many compounds in
either gaseous or particulate form, which results in a higher threat of cancer inci-
dence in children [6, 64]. Individuals who are strongly exposed to smoke, pesti-
cides, hair dye, incense including other substances or an aromatic gum may have a
higher risk of getting cancer. Certain medications and other chemical compounds
can also have carcinogenic effects as classified by IARC, such as diethylstilbestrol
used in the prevention of miscarriage, tamoxifen widely used for chemoprevention
of breast cancer and drug for chemotherapy, oxazepam, phenobarbital, and chemi-
cals like organochlorine compounds, derivatives of phenoxyacetic acids, aromatic
benzene derivatives used in the manufacture of drugs in the pharmaceutical industry
[65, 66]. Heavy metal exposure, such as lead, cadmium, arsenic, mercury, and an
extensively used persistent organochlorine pesticide endosulfan, also called as
endocrine disruptor molecules, are well known to possess oncogenic potential with
an ability to induce epigenetic alterations such as chromatin remodeling, changes in
DNA methylation profiles, modulating the expression of oncogenic enzymes as
well as the expression of microRNAs (miRNAs) and non-coding RNAs (ncRNAs),
eventually contributing to the development of various types of cancer [67–69]. A
recent study conducted in Ethiopia has shown that exposure to environmental toxi-
cants such as organochlorine pesticides might be one of the vital factors leading to
tumorigenesis. The findings highlight that several organochlorine pesticides
(dibutyl-chlorendate, endosulfan, gamma-chlordane, heptachlor, p,p’- dichlorodi-
phenyltrichloroethane, and p,p’-dichlorodiphenyldichloroethylene) were detected
in the serum of the study participants among which heptachlor, a cyclodiene insec-
ticide, was found at a higher concentration in breast cancer patients, suggesting that
organochlorine pesticides could be one of the prime risk factors for breast cancer
incidences in Ethiopia [70].
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1443
3.3 Biological Factors
Poor dietary habits and sedentary lifestyles play a deleterious role by adversely
affecting the normal health of individuals and are closely associated with cancer
prevalence. Diet, obesity, and physical inactivity are some of the factors which con-
tribute to tumor progression and are accountable for nearly 30–35% of cancer deaths
[81]. A population-based study performed on more than five million adults of
United Kingdom established that higher body mass index (BMI) was directly linked
to the progression of many types of cancer and responsible for nearly 12,000 cases
every year [82]. Prolonged sitting increases the risk of prostate, ovarian, endome-
trial and colorectal cancer and contributes significantly to cancer mortality, espe-
cially in females. Some specific food influences development of specific cancers,
for example, chewing of areca nut may cause oral cancer; a common dietary con-
taminant aflatoxin B1 causes cancer of liver, whereas diet containing high salt con-
centration is linked to stomach (gastric) cancer [74, 83, 84]. Low intake of fruits,
vegetables, and overeating of red or processed meat and the consumption of barbe-
cued or grilled fish and meat may increase the risk of pancreatic, colon and breast
cancer [9, 85]. Dietary differences between populations may have an impact on
cancer incidence. Among the Japanese population, gastric cancer is more common
1444 M. S. Ghufran et al.
because of a high salt diet, whereas colon cancer is more prevalent in the United
States population due to the consumption of more snacks containing refined
sugar [86].
4.3.2 Epigenetic Markers
4.4 Screening
Unlike diagnostic efforts driven by medical signs and symptoms, the screening pro-
cess involves solid efforts to detect cancer once it has developed but before the
appearance of any clear sign and symptoms. For many cancer types, the screening
process is inefficient, not available and generally are more complicated and requires
dedicated person and special equipment. The widespread use of mammography and
screening for prostate cancer are prime examples showing the potential pitfalls of
detection and subsequent redundancy of indolent lesions following overtreatment
[88]. People at high risk can be identified by selective screening such as the knowl-
edge about family history and background. Various factors must be considered to
ensure that the benefits of screening do not outweigh the risk of screening. A screen-
ing program must be cost-effective for the target population and must be accessible
and acceptable. In order to avoid a false positive outcome of screening programs,
the selection of patients should be based on risk factors and age. Examples of
screening methods for cervical cancer include PAP cytology test, HPV testing and
visual inspection with acetic-acid (VIA), whereas breast cancer can be screened by
mammography in setup with a strong or relatively strong health care system [22, 87,
89]. In the future, the emphasis should be on early detection and cancer prevention
efforts involving precise cancer prevention strategies so that screening and cancer
prevention programs could be matched to individual’s cancer risk attributable to
known genomic and environmental factors [88].
Cancer is a major public health issue and a costly disease in terms of treatment and
cure in both developed and developing countries. Accurate diagnosis of a particular
cancer type is critical for an appropriate and effective treatment strategy because
each type of cancer requires a precise treatment regimen. There are many treatment
options available for existing cancer types. The classical treatment methods include
surgery, hormonal therapy, radiation therapy and chemotherapy, whereas advanced
ones include image-guided radiation therapy, intensity modulated radiotherapy,
1448 M. S. Ghufran et al.
gene therapy, personalized therapy, targeted therapy, and bone marrow transplanta-
tion. Advanced treatment models are more expensive and are not readily available
in low- and middle-income countries [90, 91]. However, they have fewer side effects
with effective results. The choice of a particular treatment model depends on several
parameters, such as the types and stage of cancer, location, grade, and biology of
tumor as well as the preferences of the patient and his/her health status. The cost of
cancer treatment is very expensive in low- and middle-income countries because of
the unaffordable prices of drugs for therapy and limited access to novel treatments
[92]. The cost-effectiveness of a drug or regimen varies between different settings;
for example, some of the drugs that are economical in developed countries remain
unaffordable in low- and middle-income countries. There has been a sharp increase
in cancer drug spending forecasts, but access remains a problem. Therefore, it is
essential to address this issue by increasing uniform global availability, affordabil-
ity, and accessibility to vital cancer drugs within the reach of patients and protecting
them from financial loss, so that the fight against cancer prevention and treatment is
not left behind [93].
Sometimes, after trying many types of available treatments or even providing the
best care to cancer patients, cancer continues to spread and reaches an advanced
stage where it could be a very tough situation for the patient, family members, and
the medical doctor to talk and decide about discontinuing treatment for the cancer
and focusing on end-of-life or hospice care [94]. Palliative care is a treatment, rather
than a cure, to relieve pain, symptoms and stress caused by cancer and helps in
improving the quality of life for patients and that of their relatives by the detailed
assessment and management of the psychosocial, spiritual and physical distress
experienced by patients. Unlike treatment that aims to kill cancer cells directly, the
prime goal of palliative care is to help and support people to live more comfortably
and is especially needed in advanced stage cancer patients where there is little or no
hope of getting cured [94]. Palliative care is required in a wide range of diseases and
among adults with chronic diseases, 34% of the world’s cancer patients need pallia-
tive care. According to the WHO, 78% of the estimated 40 million people living in
low- and middle-income countries are in need of palliative care each year and, at the
same time, the global requirement will continue to rise due to the ageing of popula-
tions and the growing burden of non-communicable diseases including cancer [95].
5 Conclusion
One of the biggest challenges the world is facing today in fighting cancer is the lack
of cost-effective solutions, as the entire treatment range from classical to modern
methods is not affordable. To prevent and control cancer-related morbidity and mor-
tality, we need to strengthen and enhance public health systems, especially in low-
and lower-middle-income countries. Now is the time to invest in affordable cancer
1450 M. S. Ghufran et al.
References
1. GBD 2015 Mortality and Causes of Death Collaborators (2016) Global, regional, and national
life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death,
1980–2015: a systematic analysis for the Global Burden of Disease Study 2015 [published
correction appears in Lancet. 2017;389(10064):e1]. Lancet 388(10053):1459–1544. https://
doi.org/10.1016/S0140-6736(16)31012-1
2. Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M et al (2020) Global cancer observa-
tory: cancer today. International Agency for Research on Cancer, Lyon. Available from: https://
gco.iarc.fr/today. Accessed 10 Jan 2022
3. Ferlay J, Ervik M, Lam F, Colombet M, Mery L, Piñeros M et al (2020) Global cancer obser-
vatory: cancer tomorrow. International Agency for Research on Cancer, Lyon. Available from:
https://gco.iarc.fr/tomorrow. Accessed 10 Jan 2022
4. Anand P, Kunnumakkara AB, Sundaram C, et al (2008) Cancer is a preventable disease that
requires major lifestyle changes [published correction appears in Pharm Res 2008;25(9):2200.
Kunnumakara, Ajaikumar B [corrected to Kunnumakkara, Ajaikumar B]]. Pharm Res
25(9):2097–2116. https://doi.org/10.1007/s11095-008-9661-9
5. Budnik LT, Adam B, Albin M et al (2018) Diagnosis, monitoring and prevention of exposure-
related non-communicable diseases in the living and working environment: DiMoPEx-project
is designed to determine the impacts of environmental exposure on human health. J Occup
Med Toxicol 13:6. https://doi.org/10.1186/s12995-018-0186-9
6. Lewandowska AM, Rudzki M, Rudzki S, Lewandowski T, Laskowska B (2019) Environmental
risk factors for cancer - review paper. Ann Agric Environ Med 26(1):1–7. https://doi.
org/10.26444/aaem/94299
7. Murray CJL, Lopez AD (eds) (1996) The global burden of disease: a comprehensive assess-
ment of mortality and disability from diseases, injuries, and risk factors in 1990 and projected
to 2020. Harvard University Press, Cambridge, MA
8. Stewart BW, Kleihues P (2003) World cancer report. IARC Press, Lyon
9. Sung H, Ferlay J, Siegel RL et al (2021) Global cancer statistics 2020: GLOBOCAN esti-
mates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin
71(3):209–249. https://doi.org/10.3322/caac.21660
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1451
10. Siegel RL, Miller KD, Fuchs HE, Jemal A (2021) Cancer statistics, 2021 [published correc-
tion appears in CA Cancer J Clin 2021;71(4):359]. CA Cancer J Clin 71(1):7–33. https://doi.
org/10.3322/caac.21654
11. Jayasekara H, MacInnis RJ, Room R, English DR (2016) Long-term alcohol consumption and
breast, upper aero-digestive tract and colorectal cancer risk: a systematic review and meta-
analysis. Alcohol Alcohol 51(3):315–330. https://doi.org/10.1093/alcalc/agv110
12. de Martel C, Georges D, Bray F, Ferlay J, Clifford GM (2020) Global burden of cancer attrib-
utable to infections in 2018: a worldwide incidence analysis. Lancet Glob Health 8(2):e180–
e190. https://doi.org/10.1016/S2214-109X(19)30488-7
13. Wild CP, Weiderpass E, Stewart BW (eds) (2020) World cancer report. Cancer research for
cancer prevention. International Agency for Research on Cancer, Lyon. Available from: http://
publications.iarc.fr/586. Accessed Jan 2022
14. Wilson L, Bhatnagar P, Townsend N (2017) Comparing trends in mortality from cardiovascu-
lar disease and cancer in the United Kingdom, 1983-2013: joinpoint regression analysis. Popul
Health Metrics 15(1):23. https://doi.org/10.1186/s12963-017-0141-5
15. Omran AR (1971) The epidemiologic transition. A theory of the epidemiology of population
change. Milbank Mem Fund Q 49(4):509–538
16. Gersten O, Wilmoth JR (2002) The cancer transition in Japan since 1951. Demogr Res
7:271–306
17. Global Health Observatory (2016) World Health Organization, Geneva. Available from: http://
www.who.int/gho/database/en/. Accessed Jan 2022
18. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A (2018) Global cancer statis-
tics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185
countries [published correction appears in CA. Cancer J Clin 2020;70(4):313]. CA Cancer J
Clin 68(6):394–424. https://doi.org/10.3322/caac.21492
19. NCD Countdown 2030 collaborators (2018) NCD countdown 2030: worldwide trends in non-
communicable disease mortality and progress towards sustainable development goal target
3.4. Lancet 392(10152):1072–1088. https://doi.org/10.1016/S0140-6736(18)31992-5
20. World Health Organization (2020) WHO report on cancer: setting priorities, investing wisely
and providing care for all. World Health Organization, Geneva. Licence: CC BY-NC-SA 3.0 IGO
21. Budreviciute A, Damiati S, Sabir DK et al (2020) Management and prevention strategies for
non-communicable diseases (NCDs) and their risk factors. Front Public Health 8:574111.
https://doi.org/10.3389/fpubh.2020.574111
22. World Health Organization (2021) Cancer fact shee. Available from: https://www.who.int/
news-room/fact-sheets/detail/cancer. Accessed Jan 2022
23. Human Development Index (2020) United Nations Development Programme, New York.
Available from: http://hdr.undp.org/en/content/human-development-index-hdi. Accessed
Jan 2022
24. Bray F, Jemal A, Grey N, Ferlay J, Forman D (2012) Global cancer transitions according
to the Human Development Index (2008-2030): a population-based study. Lancet Oncol
13(8):790–801. https://doi.org/10.1016/S1470-2045(12)70211-5
25. Lam CG, Howard SC, Bouffet E, Pritchard-Jones K (2019) Science and health for all children
with cancer. Science 363(6432):1182–1186. https://doi.org/10.1126/science.aaw4892
26. CI5plus (2020) Cancer incidence in five continents time trends. International Agency for
Research on Cancer, Lyon. Available from: http://ci5.iarc.fr/CI5plus/Pages/graph1_sel.aspx.
Accessed Jan 2022
27. Allemani C, Matsuda T, Di Carlo V et al (2018) Global surveillance of trends in cancer sur-
vival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diag-
nosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet
391(10125):1023–1075. https://doi.org/10.1016/S0140-6736(17)33326-3
28. Bryere J, Dejardin O, Launay L et al (2018) Socioeconomic status and site-specific cancer
incidence, a Bayesian approach in a French Cancer Registries Network study. Eur J Cancer
Prev 27(4):391–398. https://doi.org/10.1097/CEJ.0000000000000326
1452 M. S. Ghufran et al.
49. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T (2013) UV radiation and the skin. Int J Mol Sci
14(6):12222–12248. Published 2013 Jun 7. https://doi.org/10.3390/ijms140612222
50. Stawczyk M, Łakis A, Ulatowska A, Szczerkowska-Dobosz A (2011) Evaluation and com-
parison of the risk of sunbathing addiction among selected population of women. Dermatol
Rev 98:305–311
51. Yadav VK, Awasthi P, Kumar A (2019) Detection of UV-induced thymine dimers. Methods
Mol Biol 2031:313–322. https://doi.org/10.1007/978-1-4939-9646-9_17
52. Lesiak A, Słowik-Rylska M, Kozłowski W et al (2009) Epidermal proliferation and intracel-
lular adhesion impairment as one mode of action of ultraviolet-B radiation. Adv Dermatol
Allergol XXVI(4):180–185
53. Rehm J, Gmel GE Sr, Gmel G et al (2017) The relationship between different dimensions
of alcohol use and the burden of disease-an update. Addiction 112(6):968–1001. https://doi.
org/10.1111/add.13757
54. GBD 2019 Risk Factors Collaborators (2020) Global burden of 87 risk factors in 204 coun-
tries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study
2019. Lancet 396(10258):1223–1249. https://doi.org/10.1016/S0140-6736(20)30752-2
55. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans (2012) Personal
habits and indoor combustions. Volume 100 E. A review of human carcinogens. IARC Monogr
Eval Carcinog Risks Hum 100(Pt E):1–538
56. Rumgay H, Shield K, Charvat H et al (2021) Global burden of cancer in 2020 attributable to
alcohol consumption: a population-based study. Lancet Oncol 22(8):1071–1080. https://doi.
org/10.1016/S1470-2045(21)00279-5
57. Manthey J, Shield KD, Rylett M, Hasan OSM, Probst C, Rehm J (2019) Global alco-
hol exposure between 1990 and 2017 and forecasts until 2030: a modelling study. Lancet
393(10190):2493–2502. https://doi.org/10.1016/S0140-6736(18)32744-2
58. World Health Organization (2017) WHO report on the global tobacco epidemic, 2017: moni-
toring tobacco use and prevention policies. World Health Organization. https://apps.who.int/
iris/handle/10665/255874. License: CC BY-NC-SA 3.0 IGO
59. Reitsma MB, Flor LS, Mullany EC, Gupta V, Hay SI, Gakidou E (2021) Spatial, temporal, and
demographic patterns in prevalence of smoking tobacco use and initiation among young peo-
ple in 204 countries and territories, 1990-2019. Lancet Public Health 6(7):e472–e481. https://
doi.org/10.1016/S2468-2667(21)00102-X
60. GBD 2019 Tobacco Collaborators (2021) Spatial, temporal, and demographic patterns in prev-
alence of smoking tobacco use and attributable disease burden in 204 countries and territories,
1990–2019: a systematic analysis from the Global Burden of Disease Study 2019 [published
correction appears in Lancet. 2021;397(10292):2336]. Lancet 397(10292):2337–2360. https://
doi.org/10.1016/S0140-6736(21)01169-7
61. World Health Organization (2011) WHO report on the global tobacco epidemic, 2011:
warning about the dangers of tobacco. World Health Organization. https://apps.who.int/iris/
handle/10665/44616
62. Siddiqi K, Husain S, Vidyasagaran A, Readshaw A, Mishu MP, Sheikh A (2020) Global burden
of disease due to smokeless tobacco consumption in adults: an updated analysis of data from
127 countries. BMC Med 18(1):222. https://doi.org/10.1186/s12916-020-01677-9
63. World Health Organization (2021) India loses 1% of its GDP to diseases and early deaths
from tobacco use, finds WHO study. Available from: https://www.who.int/india/news/
detail/09-02-2021-india-loses-1-of-its-gdp-to-diseases-and-early-deaths-from-tobacco-use-
finds-who-study. Accessed Jan 2022
64. Fung TT, Hu FB, Hankinson SE, Willett WC, Holmes MD (2011) Low-carbohydrate diets,
dietary approaches to stop hypertension-style diets, and the risk of postmenopausal breast
cancer. Am J Epidemiol 174(6):652–660. https://doi.org/10.1093/aje/kwr148
65. O’Mahony M, Hegarty J (2009) Help seeking for cancer symptoms: a review of the literature.
Oncol Nurs Forum 36(4):E178–E184. https://doi.org/10.1188/09.ONF.E178-E184
1454 M. S. Ghufran et al.
66. IARC Monographs on the Evaluations of Carcinogenic Risks to Humans (2011) A review of
human carcinogens. Part A: pharmaceuticals, vol 100. IARC, Lyon
67. Wallace DR, Taalab YM, Heinze S et al (2020) Toxic-metal-induced alteration in miRNA
expression profile as a proposed mechanism for disease development. Cell 9(4):901. https://
doi.org/10.3390/cells9040901
68. de Araújo ML, Gomes BC, Devóz PP et al (2021) Association between miR-148a and DNA
methylation profile in individuals exposed to lead (Pb). Front Genet 12:620744. https://doi.
org/10.3389/fgene.2021.620744
69. Singh VK, Pal R, Srivastava P, Misra G, Shukla Y, Sharma PK (2021) Exposure of androgen
mimicking environmental chemicals enhances proliferation of prostate cancer (LNCaP) cells
by inducing AR expression and epigenetic modifications. Environ Pollut 272:116397. https://
doi.org/10.1016/j.envpol.2020.116397
70. Mekonen S, Ibrahim M, Astatkie H, Abreha A (2021) Exposure to organochlorine pesticides
as a predictor to breast cancer: a case-control study among Ethiopian women [published cor-
rection appears in PLoS One. 2021;16(11):e0260106]. PLoS One 16(9):e0257704. https://doi.
org/10.1371/journal.pone.0257704
71. Ksouri R (2019) Food components and diet habits: chief factors of cancer development. Food
Qual Saf 3(4):227–231. https://doi.org/10.1093/fqsafe/fyz021
72. Bbosa GS, Kitya D, Odda J et al (2013) Aflatoxins metabolism, effects on epigenetic mech-
anisms and their role in carcinogenesis. Sci Res 5(10A):14–34. https://doi.org/10.4236/
health.2013.510A1003
73. Bräse S, Encinas A, Keck J, Nising CF (2009) Chemistry and biology of mycotoxins and
related fungal metabolites. Chem Rev 109(9):3903–3990. https://doi.org/10.1021/cr050001f
74. Hamid AS, Tesfamariam IG, Zhang Y, Zhang ZG (2013) Aflatoxin B1-induced hepatocellular
carcinoma in developing countries: geographical distribution, mechanism of action and pre-
vention. Oncol Lett 5(4):1087–1092. https://doi.org/10.3892/ol.2013.1169
75. Massey TE, Smith GB, Tam AS (2000) Mechanisms of aflatoxin B1 lung tumorigenesis. Exp
Lung Res 26(8):673–683. https://doi.org/10.1080/01902140150216756
76. Boonen J, Malysheva SV, Taevernier L et al (2012) Human skin penetration of selected model
mycotoxins. Toxicology 301(1–3):21–32. https://doi.org/10.1016/j.tox.2012.06.012
77. Soni P, Ghufran MS, Olakkaran S, Puttaswamygowda GH, Duddukuri GR, Kanade SR (2021)
Epigenetic alterations induced by aflatoxin B1: an in vitro and in vivo approach with empha-
sis on enhancer of zeste homologue-2/p21 axis. Sci Total Environ 762:143175. https://doi.
org/10.1016/j.scitotenv.2020.143175
78. Ghufran MS, Soni P, Kanade SR (2019) Aflatoxin-induced upregulation of protein arginine
methyltransferase 5 is mediated by protein kinase C and extracellular signal-regulated kinase.
Cell Biol Toxicol 35(1):67–80. https://doi.org/10.1007/s10565-018-9439-8
79. Soni P, Ghufran MS, Kanade SR (2018) Aflatoxin B1 induced multiple epigenetic modu-
lators in human epithelial cell lines. Toxicon 151:119–128. https://doi.org/10.1016/j.
toxicon.2018.07.011
80. Ghufran MS, Ghosh K, Kanade SR (2016) Aflatoxin B1 induced upregulation of protein argi-
nine methyltransferase 5 in human cell lines. Toxicon 119:117–121. https://doi.org/10.1016/j.
toxicon.2016.05.015
81. Rock CL, Thomson C, Gansler T et al (2020) American cancer society guideline for diet
and physical activity for cancer prevention. CA Cancer J Clin 70(4):245–271. https://doi.
org/10.3322/caac.21591
82. Bhaskaran K, Douglas I, Forbes H, dos-Santos-Silva I, Leon DA, Smeeth L (2014) Body-mass
index and risk of 22 specific cancers: a population-based cohort study of 5·24 million UK
adults. Lancet 384(9945):755–765. https://doi.org/10.1016/S0140-6736(14)60892-8
83. Park JH, Moon JH, Kim HJ, Kong MH, Oh YH (2020) Sedentary lifestyle: overview of updated
evidence of potential health risks. Korean J Fam Med 41(6):365–373. https://doi.org/10.4082/
kjfm.20.0165
The Global Concern for Cancer Emergence and Its Prevention: A Systematic Unveiling… 1455
84. Park S, Bae J, Nam BH, Yoo KY (2008) Aetiology of cancer in Asia. Asian Pac J Cancer Prev
9(3):371–380
85. Zheng W, Lee SA (2009) Well-done meat intake, heterocyclic amine exposure, and cancer risk.
Nutr Cancer 61(4):437–446. https://doi.org/10.1080/01635580802710741
86. Brenner H, Rothenbacher D, Arndt V (2009) Epidemiology of stomach cancer. Methods Mol
Biol 472:467–477. https://doi.org/10.1007/978-1-60327-492-0_23
87. Shah SC, Kayamba V, Peek RM Jr, Heimburger D (2019) Cancer control in low- and middle-
income countries: is it time to consider screening? J Glob Oncol 5:1–8. https://doi.org/10.1200/
JGO.18.00200
88. Loomans-Kropp HA, Umar A (2019) Cancer prevention and screening: the next step in the
era of precision medicine. NPJ Precis Oncol 3:3. https://doi.org/10.1038/s41698-018-0075-9
89. Azene GK (2021) Visual inspection with acetic-acid (VIA) service utilization and associated
factors among women in Hawassa city, southern Ethiopia: a community based cross-sectional
study. Womens Midlife Health 7(1):6. https://doi.org/10.1186/s40695-021-00065-4
90. Gerber DE (2008) Targeted therapies: a new generation of cancer treatments. Am Fam
Physician 77(3):311–319
91. Kawakami K, Nakajima O, Morishita R, Nagai R (2006) Targeted anticancer immunotoxins
and cytotoxic agents with direct killing moieties. ScientificWorldJournal 6:781–790. https://
doi.org/10.1100/tsw.2006.162
92. Siddiqui M, Rajkumar SV (2012) The high cost of cancer drugs and what we can do about it.
Mayo Clin Proc 87(10):935–943. https://doi.org/10.1016/j.mayocp.2012.07.007
93. Ocran Mattila P, Ahmad R, Hasan SS, Babar ZU (2021) Availability, affordability, access, and
pricing of anti-cancer medicines in low- and middle-income countries: a systematic review
of literature. Front Public Health 9:628744. Published 2021 Apr 30. https://doi.org/10.3389/
fpubh.2021.628744
94. Rome RB, Luminais HH, Bourgeois DA, Blais CM (2011) The role of palliative care at the end
of life. Ochsner J 11(4):348–352
95. World Health Organization (2020) Palliative care fact sheet. Available from: https://www.who.
int/news-room/fact-sheets/detail/palliative-care. Accessed Jan 2022
96. Gupta J, Sharma S, Sharma NR, Kabra D (2020) Phytochemicals enriched in spices: a source
of natural epigenetic therapy. Arch Pharm Res 43(2):171–186. https://doi.org/10.1007/
s12272-019-01203-3
97. Dehelean CA, Marcovici I, Soica C et al (2021) Plant-derived anticancer compounds as new
perspectives in drug discovery and alternative therapy. Molecules 26(4):1109. https://doi.
org/10.3390/molecules26041109
98. Rizeq B, Gupta I, Ilesanmi J, AlSafran M, Rahman MDM, Ouhtit A (2020) The power of
phytochemicals combination in cancer chemoprevention. J Cancer 11(15):4521–4533. https://
doi.org/10.7150/jca.34374
Commonly Used Poisonous Medicinal
Plants in Unani System of Medicine
1 Introduction
Plants have been used since the dawn of time. It’s been a part of human civilization
for millennia. Plants are used in traditional medicine in Africa to cure a variety of
infectious and non-infectious illnesses. Body washes, massages, ingestions and
other treatments are among them. Plants produce a vast range of bio-chemicals that
are beneficial to humans. Food, colours, scents, agricultural chemicals and medica-
tions are all examples of their use. Traditional medicine is used by around 40–90%
of people in underdeveloped nations, according to the World Health Organization
(WHO) [1]. The usage of various portions of multiple medicinal plants to treat spe-
cific ailments has been popular in India since ancient times. For numerous centuries,
the indigenous medical systems of Ayurvedic, Siddha and Unani have existed. The
use of poisonous plants in the Unani system of medicine aids in the treatment of
certain diseases (Table 1). It is critical to be aware of dangerous plants that, when
administered in the correct, prescribed dose can be effective therapeutic agents [2].
Plant intoxication is a topic that has sparked a lot of discussion in the literature,
both in terms of human and animal poisoning (Table 2). Depending on the precise
components and metabolites that they contain, different herbs can have a variety of
impacts on human organ systems. Gastroenteritis, hepatotoxicity, peripheral neu-
ropathy, psychosis, rhabdomyolysis and blood count abnormalities are all possible
side effects, as are cardio toxicity and multisystem organ failure [3]. Poisoning can
occur through touch, which causes skin irritation; ingestion, which causes internal
S. A. A. Makbul (*)
Department of Ilmul Advia (Pharmacology), State Unani Medical College & Hospital,
Prayagraj, Uttar Pradesh, India
S. Rahman
Department of Ilmul Advia (Pharmacology), Eram Unani Medical College and Hospital,
Lucknow, Uttar Pradesh, India
Table 1 List of Poisonous drugs used in Unani system of medicine with part used, formulations
and indications
Sr. Unani Botanical Part
no. name name used Formulations Indications References
1 Ain-ud- Abrus Seed,Dawa-e-Tikor, Bahaq (Pityriasis), Bars [18, 19,
Deek/ precatorius Root Zimad-e- (Leukoderma), Kalaf 55]
Ghonchi L. Mohasa, (Freekles), Sabal
Tila-e-Aroosak, (pannus), Bayaz-e-
Raughan Ganj, Chashm (Corneal
Majoon-e- opacity) (as a Surma)
Malook,
Tila-e-Khas-ul-
Khas, Tila-e-
Jamal Gota
2 Ajwain Hyoscyamus Seed, Barsha’sha, Sahr (Insomnia), [18, 55,
Khurasani niger L. Leaf, Banadiq-ul- Malekhuliya 56, 11, 12]
Extract Buzoor, (Melancholia),
Tiryaq-e-Nazla, Waj-ul-Mafasil
Habb-ul-Misk, (rthritis), Irq-un-Nasa
Habb-e-Jadwar, (Sciatica), Naqras
Habb-e-Mumsik (Gout), Dard-edandan
tilayi, Halwa (Tootache), Dard-e-
supari pak, Gosh (Earache),
Qurs-e-Tinkar, Surat-e-Inzal (Pre-
Qurs-e-Jaryan, mature ejaculation),
Qurs-e- Jarayan-e-Mani
Musallas, (Spermatorrhoea),
Majoon-e-Jalali, Su’al-e-Yabis (Dry
Majoon cough), Ishal (Loose
Murawwih-ul- motion)
Arwaah, Majoon
muqawwi wa
mumsik
3 Azaraqi/ Strychnos Seed Habb-e-Azaraqi, Falij (Hemiplegia), [18, 55,
Kuchla nux-vomica Majoon-e- Laqwa (Facial 56, 20, 11]
L. Azaraqi, paralysis), Waj-ul-Qutn
Habb-e-Khas, (Backache), Zeeq-un-
Habb-e-Fauladi, Nafas (Breathlessness),
Safoof-e- Sil (Tuberculosis),
Azaraqi, Suzak Gonorrhoea),
Habb-e- Su’aal Muzmin
Marwareedi, (Chronic cough),
Roghan-e- Zo’f-e-Ishtiha
Kuchla, (Anorexia), Ghashi
Majoon-e-Lana, (Syncope), Behoshi
Habb-e- (Unconsciousness),
Muna’yish Zo’f-e-Qalb (Weakness
of heart), Zo’f-e-Meda
(Weakness of
stomach)), Juzam
(Leprosy)
(continued)
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1459
Table 1 (continued)
Sr. Unani Botanical Part
no. name name used Formulations Indications References
4 Afyun/ Papaver LatexHabb-e-Jadwar, Qaulanj (Colitis), [18, 55,
Afeem somniferum Barsha’sha, Dard-e-Sar (Headache), 56, 11]
L. Habb-e-Hamal, Waj-ul-Asab (Nervine
Habb-e-Surfa, pain), Wajul-Asnan
Habb-e-Sumaq, (Toothache), Sahr
Habb-e-Mumsik (Insomnia), Junoon
tilayi, Habb-e- (Insanity), Waj-ul-
Mumsik Ambari, Mafasil (Arthritis),
Habb-e-Nazla, Sarsam (Meningitis),
Roghan-e- Shahiqa (Whooping
Kuchla, cough), Su’al (Cough),
Roghan-e-Kalan, Zat-ul-janb (Pleurisy),
Shayyaf-e- Waj-ul-Qutn
Abyaz, (Backache), Surat-e-
Ques-e- Inzal (Pre-mature
Musallas, ejaculation), Nazla wa
Majoon Zukam (Cold and
muqawwi wa coryza).
Mumsik,
Habb-e-Pechish
5 Baladur/ Semecarpus Fruit, Anqaroya Zof-e-Asab [18, 20,
Bhalawan anacardium Fruit Kabeer, (Neurasthenia), 55, 56]
L. Extract Anqardia Zof-e-Hafiza (Weakness
Sagheer, of memory power),
Majoon-e- Zof-e-Qalb (Weakness
Baladur, of heart), Falij
Roghan-e- (Hemiplegia), Laqwa
Bhalawan (Facial paralysis)
6 Bed Ricinus Seed, Zimad-e- Waja-ul–Mafasil [18, 20,
anjeer/ communis L. leaf Jalinoos, (Arthritis), Falij 55, 56]
Arandi Zimad-e-Sheer- (Hemiplegia), Laqwa
e-Shutr, (Facial paralysis),
Marham-e- Deedan-e-Ama
Atishk, (Intestinal worms),
Marham-e- Ehtebas-e- Haiz
Dakhaliyoon, (amenorrhoea), Sual
Marham-e- (Cough), Ra’sha
Rusul, Tiryaq-e- (Tremor), Zeeq-un-
Pechish, Nafas (Breathlessness),
Roghan-e- Qaulanj (Colitis),
Chahar Barg Istesqa (Ascites)
(continued)
1460 S. A. A. Makbul and S. Rahman
Table 1 (continued)
Sr. Unani Botanical Part
no. name name used Formulations Indications References
7 Beesh/ Aconitum Root Habb-e-Miskeen Shaqiqa (Migrain), [18, 20,
Bachhnak napellus L. Nawaz, Qurs Irq-un Nasa (Sciatica), 55, 56]
Zat-ul-Janb (Pleurisy),
ikseer-e-Falij wa
laqwa, Zat-ur-Riya
Habb-e-Nuqra, (Pneumonia), Humma
Ikseer-e-Surkh (Fever), Ehtabas-e-Baul
(Retention of Urine),
Ehtabas-e-Haiz
(Amenorrhoea), Juzam
(Leprosy), Bars
(Lucodrema),
Zeeq-un-Nafas
(Breathlessness),
Qurooh-e-Khabisa
(Non-healing ulcer),
Waj-ul-Mafasil
(Arthritis)
8 Habb-us- Croton Seed Habb-e-Miskeen Amraz-e-Balghami wa [18, 20,
Salateen/ tiglium L. Nawaz, Sawdavi (Phlegm and 55, 56]
Jamalgota Roghan-e- black bile diseases),
Jamalgota, Daad (Ring worm),
Tila-e-Surkh, Ganj, Bars
Habb-e-Mushil, (Leucoderma),
Dawa-e-Siyah-e- Wajaul- Mafasil
Mushil (Arthritis), Qoolanj
(Colitis), Atishk
(Syphilis), Juzam
(leprosy), Istisqa
(Ascites), Qabz qawi
(Chronic constipation),
Sa’fa (Porrigo),
Da’-us-Salab
(Alopecia)
9 Jauz-e- Datura Seed, Habb-e-Shifa, Waja-ul –Mafasil [18, 20,
Masil/ metel L. Leaf Habb-e-Ziq-un- (Arthritis), Niqras 55, 56]
Dhatura Nafs, Habb-e- (Gout), Sual (Cough),
Mumsik tilayi, Zeequn Nafas
Roghan-e- (Breathlessness), Nazla
Dhatura, (Catarrh), Humma
Majoon-e-Falak (Fever), Dama
ser, Roghan-e- (Asthma), Shaheeqa
Haft-e-Barg, (Migrain), Kanphed
Tila-e-Jadeed (Mumps), Sahr
(Insomnia), Suda’
(Headache), Surat-e-
Inzal (Pre-mature
ejaculation)
(continued)
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1461
Table 1 (continued)
Sr. Unani Botanical Part
no. name name used Formulations Indications References
11 Qinnab/ Cannabis Whole Habb-e-Hamal, Ishal (Loose motion), [18, 20,
Bhang sativa L. Plant, Habb-e-Mumsik Kasrat-e-Tams 55, 56]
Leaf (Menorrhagia),
tilayi, Habb-e-
Mumsik Ambari,Bawaseer (Piles), Sual
Majoon Falak (Cough), Waj-ul-Kabid
(Liver pain), Qulanj
ser, Majoon-e-
Muqawwi wa (Colitis), Sahr
Mumsik, (Insomnia), Junoon
Safof-e-Kalan,(Insanity), Shaqeeqa
Majoon-e- (Migrain), Suda-e-
Muqawwi alvi Dayimi (Chronic
Khan, Majoon-Headache), Kuzaz
e-Mubahhi (Tetanus), Syrat-e-Inzal
antaki (Pre-mature
ejaculation), Shaheeqa
(Whooping cough)
12 Tambak/ Nicotiana Leaf Bakhoor-e- Waj-ul-Asnan [55, 56]
Tambaku tobacum L. Damah, (Toothache), Warm-e-
Sanoon-e- Lussa (Gum
Tambakoo, inflammation), Dard sar
Marham-e- bawajh-e-zukam
Muhallil (Headache due to cold),
Warm-e-Khusyatain
(Orchitis)
13 Ushr/ Calotropis Root, Habb-e-Seen, Zeequn Nafas [18, 55,
Madar procera Bark, Habb-e-Gul-e- (Breathlessness), 56]
(Ait) R.Br. Leaf, Aakh, Roghan- Waj-ul-Mafasil
Seed, e-Auja’-e-Khas, (Arthritis), Bawaseer
Flower Roghan-e-Surhk, (Piles), Zaheer
Raghan-e-Gul-e- (dysentry) Deedan-e-
Aakh, Qurs-e- Ama (Intestinal
Gul-e-Aakh, worms), Zof-e-Meda
Roghan-e- (Weakness of stomach),
Chahar Barg, Dad (Ring worm),
Roghan-e-Auraq Dama (Asthma), Su’al
(Cough), Musqit-e-
Janeen (abortificient),
Kharash-e-Meda
(Roughness of stomach)
poisoning; or absorption, which causes internal poisoning [2]. A wide range of phy-
tochemicals, such as alkaloids and glycosides, are derived from plant sources and
are useful in the treatment of a variety of illnesses. These phytochemicals have anti-
allergy, anti-inflammatory, purgative, laxative and anti-migraine properties (Table 3)
[4]. There are various plant species that are poisonous or harmful to humans. Plant
toxins have a variety of beneficial effects and can be utilized to cure a variety of ail-
ments. They can be tweaked to improve affinity and effectiveness. Medicinal plants
1462 S. A. A. Makbul and S. Rahman
Table 2 (continued)
Sr.
no. Botanical name Signs and symptoms References
5 Croton tiglium Croton tiglium L. is an irritant poison and a drastic purgative [59]
L. drug; its strong cathartic action begins with irritation in
stomach. The mucosa of the gastrointestinal tract has a
tendency to become hypersensitive to Croton tiglium L.,
resulting in symptoms such as searing pain from the mouth
to the oesophagus, nausea, vomiting, bloody faeces and
dehydration, which can lead to a weak pulse, loss of body
temperature and death..
6 Semecarpus The powerful irritant properties of the juice of pericarp have [31]
anacardium L. frequently been made use of by malingerers in producing
ophthalmic and skin lesions and also in producing abortions.
Dermatitis can develop in persons who prepare the oil, apply
it to garments in their job as a laundryman, or wear the
marked clothing. Skin lesions and anuria developed after
exposure to S. anacardium sap, followed by diffused cortical
necrosis. External application of S. anacardium caused
painful micturition, with urine that was scarlet and bloody
and stools that were very painful to pass.
7 C. procera The latex from the plant has used by the tribal people to [33]
(Ait.) R.Br. make poison arrows used for hunting purpose. The latex is
aand Calotropis highly toxic to human eyes cause ocular toxicity and
gigantean (L.) produces loss of vision with photophobia. Latex of C.
R.Br procera was studied for its inflammatory effects using pedal
oedema and air pouch models of inflammation in rats and
could be used to evaluate anti-inflammatory drugs.
Furthermore, latex also produces toxic iridocyclitis,
keratoconjunctivitis, corneal endothelial cytotoxicity and
keratitis when applied accidentally on the eye.
8 Aconitum Spp. In recent years, there have been a few reports of [38, 53]
cardiotoxicity and neurotoxicity caused by insufficient
aconitine ingestion. Toxic effects mostly impact the central
nervous system and heart, with gastrointestinal symptoms
present as well. The development of ventricular
tachyarrhythmia and cardiac arrest was the cause of death.
9 Cannabis sativa Short-term use of cannabis has led to impaired short-term [60]
L. memory; impaired motor coordination; altered judgment;
and paranoia or psychosis at high doses. Addiction, altered
brain development, cognitive impairment, poor educational
outcomes (e.g. dropping out of school) and lower life
satisfaction have all been linked to long-term or excessive
cannabis use, particularly in teens. In people who are
predisposed to such problems, long-term or excessive
cannabis usage is linked to chronic bronchitis and an
increased risk of chronic psychosis-related health
conditions, such as schizophrenia and depressive variations.
Cannabis use has also been linked to vascular disorders such
as myocardial infarction, stroke and transient ischemic
attack.
(continued)
1464 S. A. A. Makbul and S. Rahman
Table 2 (continued)
Sr.
no. Botanical name Signs and symptoms References
10 Nicotiana Nicotine is one of the most deadly poisons available, with a [46]
tobacum L. quick onset of action. The peripheral and central nervous
systems are the target organs, in addition to local activities.
Nicotine produces irritation and burning in the mouth and
throat in humans, as well as increased salivation, nausea,
abdominal pain, vomiting and diarrhoea. The effects on the
gastrointestinal tract are less severe, but they might still
occur following cutaneous and respiratory exposure.
Increased blood pressure and pulse rate. Nicotine also
causes an increase in plasma free fatty acids, hyperglycemia
and a rise in catecholamine levels in the bloodstream. The
blood supply to the heart is lowered, but the blood flow to
the skeletal muscles is enhanced. Tremors, prostration,
cyanosis, dyspnoea, convulsions and collapse are all
symptoms of severe poisoning.
that are poisonous are used to treat a variety of diseases. The Unani school of medi-
cine employs medications derived from plants, animals and minerals, some of
which have toxic ingredients and are even lethal poisons like Semiecarpus anacar-
dium L., Abrus precatorius, Ricinus communis, Strychnos nuxomica etc., Therefore,
these drugs are rectified to minimize their toxicity [5]. Medicinal plants are fre-
quently used and thought to be harmless, yet they can also be harmful. When medi-
cal plant poisoning has been documented, it is usually due to mistaken identity in
the form in which they have been sold, or faulty preparation by untrained persons.
The medications become physically and chemically pure after the purifying pro-
cess, making them more therapeutically effective and less harmful. The purpose of
this chapter is to highlight the toxicity of medications, chemical ingredients, detoxi-
fication methods and pharmacological features of hazardous medicinal plants
employed in the Unani system of medicine.
Strychnos nux-vomica L.
1.1
Sr. Botanical
no. Unani name name Chemical constituents Pharmacological activities References
5 Jauz-e-Masil / Datura Atropine, scopolamine, phenolic compounds, tannins, Antiarthritic, antifungal, antistress, [23, 50,
Dhtura stramoniumflavonoids, cardiac glycosides, alanine, phenylalanine, antiulcer, hypoglycaemic, anticancer, 51]
glutamate, tyrosine, tigloidin, aposcopolamine, apoatropin, antiasthmatic, antimicrobial, anti-
hyoscyamine N-oxide, scopolamine N-oxide17–20, inflammatory, Anticholinergic,
ditigloyloxytropane, 7-hydroxyhyoscyamine Larvicidal, mosquito repellent,
6 Habb-us-Salateen/ Croton alkaloids, flavonoids, saponins, linoleic acid, oleic acid, Antitumour, antioxidant, [25, 52]
Jamalgota tiglium L. myristic acid, tigliane phorbol esters, 13-O-acetylphorbol-20- antidermatophytic, anticonvulsant
linoleate, 13-O-ttigloylphorbol-20-linoleate,
12-oacetylphorbol-13-tigliate, 12-O-decanoylphorbol-13-(2-
methylbutirate) and 12-O-acetlyphorbol-13-decanoate
7 Baladur/Bhalawan Semecarpus Bhilavanol A (monoenepentadecyl catechol I), bhilavanol B Analgesic, Hypoglycemic, [30, 31]
anacardium (dienepentadecyl catechol II), biflavones A, C, A1, A2; Hepatoprotective, Anthelmintic,
L. tetrahydrorobustaflavone, tetrahydroamentoflavone, Anti-Cancer, Anti-Inflammatory,
jeediflavone, jeediflavanone, semecarpuflavanone, Neuroprotective, Antimicrobial,
galluflavanone, amentoflavanone, nallaflavanone, Anti-Spermatogenic, Memory
semicarpetin, anacarduflavanone, otrimethyl bioflavanone Enhancing, Cardioprotective,
A1, o-trimethyl bioflavanone A2, o-tetramethyl bioflavanone Aphrodisiac, Anti-Tuberculosis,
A1, o-hexamethl bichalcone A, o-dimethyl biflavanone B, Anti-arthritic, Antitumour,
o-heptamethyl bichalcone B1, o-hexamethyl bichalcone B2, Antineoplastic, Cytotoxic, Cytostatic,
o-tetramethyl biflavanone C, anacardic acid, cardol, catechol,
alkenyl catechols, fixed oil, semecarpetin, anacardol,
anacardoside, semecarpol. Monolefin I, diolefin II, oleic acid,
linoleic acid, palmitic acid, stearic acid and arachidic acid.
S. A. A. Makbul and S. Rahman
Sr. Botanical
no. Unani name name Chemical constituents Pharmacological activities References
8 Ushr/Madar C. pprocera Calotropin, calotropagenin, calotoxin, calactin, uscharin, Acaricidal, schizonticidal, [32, 33,
(Ait.) R.Br. amyrin, amyrin esters, uscharidin, coroglaucigenin, antimicrobial, Antifertility, 34]
aand frugoside, corotoxigenin, calotropagenin, voruscharine. The anthelmintic, insecticidal, anti-
Calotropis latex contains fatty acids terpenels, sterols and hydrocarbons. inflammatory, antidiarrheal,
gigantean Lead, zinc, cadmium, nickel, calotoxin, iron, copper were Antimalarial, Gastroprotective,
(L.) R.Br identified in the latex and leaves of calotropis. Calcium anticancerous, larvicidal, Analgesic,
present highest in leaves and magnes in latex. Root barks antinociceptive, antipyretic,
contain calotropterpenyl, mundarol isovalerate and Antibacterial, antiparasitic
rutinoside. antimicrobial, antioxidant,
antiasthmatic, Anticancer
9 Afyun/Afeem Papaver Morphine, codein, neopine, thebain, meconic acid, Lactic, Antiallodynic, analgesics, antitussive, [35]
somniferum Malic, Tartaric acid, Citric acid, Acetic acid, Succinic acid, anti-diarrheal, anti- smoking,
L. sulphuric acid, Phosphoric acid, calcium, phosphorus, antidepressant, anti-anxiety,
sodium, magnesium, etc., Protopine, cryptopine, Benzyl bronchodilator, antibacterial, narcotic
isoquonoline,
10 Beesh/Bachhnak Aconitum benzoylmesaconine, mesaconitine (MA), AC, hypaconitine Anti-inflammatory, analgesia, [38, 53]
Spp. (HA), heteratisine, heterophyllisine, heterophylline, anti-rheumatism, cardiotonic actions
heterophyllidine, atidine, isotisine, hetidine, hetsinone,
benzoylheteratisine. Quercetin 7-O-(6-transcaffeoyl)-β-
glucopyranosyl-(1 → 3)-α-rhamnopyranoside-3-O-β-
glucopyranoside, kaempferol
7-O-(6-trans-caffeoyl)-β-
glucopyranosyl-(1 → 3)-α-rhamnopyranoside-3-O-β-
gglucopyranoside and kaempferol
7-O-(6-trans-p-coumaroyl)-β-
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine
glucopyranosyl-(1 → 3)-α-rhamnopyranoside-3-O-β-
glucopyranoside, together with β-3,4-dihydroxyphenethyl
β-glucopyranoside. Palmitic acid, oleic acid
(continued)
1467
Table 3 (continued)
1468
Sr. Botanical
no. Unani name name Chemical constituents Pharmacological activities References
11 Qinnab/Bhang Cannabis Tetrahydrocannabinol, Cannabidiol, Cannabidiol, Antidiabetic, anticancer, Parkinson’s [42]
sativa L. Cannabinol, β-caryophyllene, Cannabigerol disease, Chronic pain and arthritis,
Nausea vomiting
12 Tambaku Nicotiana Nicotine, nicoteine, nicotimine, anabaine anatalline, Antibacterial, anti-nociceptive, [54]
tabacum L. nornicotine, glucosides, tahacinin, tahacilin and isoquercitrin, antimicrobial, antifungal, anhelmintic,
1-quinic, chlorogenic, caffeic, oxalic acids, anatabine, Anti Alzheimer’s,
nornicotine, Quercetin-3,3′-dimethyl ether, gibberellins-
nicotiana α, β and γ,
S. A. A. Makbul and S. Rahman
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1469
hairs radiating from the centre to the sides, thus giving the seed a very characteristic
sheen. The seed with dark grey horny endosperm, where the small embryo is housed,
has no odour but tastes very bitter [8]. In several southern Asian countries, nux-
vomica is commonly used as a medication. Although brucine has a promising phar-
macological profile, its therapeutic application is hampered by substantial central
nervous system toxicity. The therapeutic window for brucine is relatively small;
LD50 of 50.10 mg/kg was recorded [7]. Detoxified Azaraqi (seed) used in Unani
medicine for various ailments of brain and nerves. Due to its nerve tonic and nerve
stimulant properties, it is used for the treatment of paralysis, bell’s palsy, chorea,
arthritis etc. [6].
1.1.1 Detoxification/Rectification Methods
Azaraqi 70 g is buried in Multani mitti (Fuller’s earth) and water is poured over it
daily for 10 days, then it is removed and washed. After removing the embryo por-
tion, the outer covering is peeled off using a knife, and the cotyledons are separated.
Subsequently it is washed with hot water and tied it in a clean cloth bag. The bag is
immersed in a vessel containing two litre of milk. The milk is then boiled till it
evaporates; care is taken that the bag does not touch the bottom of the vessel.
Thereafter, it is removed from the bag and washed with water to obtain Azaraqi
Mudabbar [5, 9, 10, 11].
Hyoscyamus niger L.
1.2
The drug Ajwain khurasani consists dried seed of Hyoscyamus niger L.(Family:
Solanaceae), a small tree found in Pakistan and other Asia’s countries, mostly in
Khurasan, a city of Iran; in India it is found in Akbrabad and Himalayan (Kashmir
to Gadhwal). The fresh herb smells as of Ajwain (Trachyspermum ammi), for this
reason called Ajwain Khurasani [9, 12]. Henbane means ‘hen killer’ in English, and
its name comes from the Anglo-Saxon words hen (chicken) and bana (murderer),
because fowls that ingest its seeds become paralysed and die. Black henbane is an
annual or biennial plant that can reach a height of three feet. Hundreds of tiny black
seeds, 1.5 mm long, are in egg-shaped fruit. One plant produces about 10,000 seeds
[13]. Its seeds are reniform or discoid, minutely pitted; embryo ringlike or
coiled [14].
1470 S. A. A. Makbul and S. Rahman
Abrus precatorius L.
1.3
Ricinus communis
1.4
The drug Bed Anjeer consists of mature and dried seeds of Ricinus communis
L. (Family: Euphorbiaceae). Popularly known as ‘castor plant‘a tall glabrous shrub
or almost small tree, 2–12 m high, found throughout India, mostly growing wild on
waste land and also cultivated for its oil seeds [18–20]. The plant is widespread
throughout tropical regions as ornamental plants. The castor oil plant is a fast-
growing, suckering perennial shrub or occasionally a soft wooded small tree up to
6 m or more, but it is not hardy in nature. The leaves are green or reddish in colour
and have a diameter of 30–60 cm. The leaves are alternating and palmate, with 5–12
deep lobes with coarsely serrated segments. The monoecious flowers are 30–60 cm
long. The fruit is a three-celled thorny capsule. The capsule of fruit covered with
soft spins like processes and dehiscing in to three 2-valved cocci [21]. The seeds are
considerable differences in size and colour. Seeds rectangular, one face convex, the
other slightly flattened, 1–1.5 cm long, 0.6–0.9 cm wide, 0.4–0.8 cm thick. The testa
hard, glossy, smooth, grey or brown to red-dish-brown or black, and may be mar-
bled or striped, raphe runs from caruncle to chalaza. Its taste weakly acrid [19, 21].
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1471
Seeds are the primary source of oil which is in use both as an herbal medicine and
as a conventional therapy for various ailments [22].
Croton tiglium L.
1.6
1.6.1 Detoxification/Rectification Methods
Kernels of the C. tiglium L. 25 g are tied in a cloth bag and boiled in one litre of
Cow’s milk till the milk becomes dense. The kernels are then removed from the bag
and the embryo part of the seeds is removed to obtain Jamalgota Mudabbar when
they have cooled [28, 29, 10].
1472 S. A. A. Makbul and S. Rahman
Semecarpus anacardium L.
1.7
1.7.1 Detoxification/Rectification Methods
With the use of red hot tongs, the juicy contents (Asal-e-Bhilawan) are squeezed out
fully after the fruit‘s cap (thalamus) is removed. After that, the fruits are boiled three
times in freshwater. Finally, the fruits are cooked in milk, rinsed and dried.
Precaution must be taken not to touch the juice with hands as the juice is toxic
[5, 9, 10].
Calotropis procera and Calotropis gigantean (Family: Apocynaceae) are two com-
mon species of calotropis. Calotropis is a perennial plant and is a genus of flowering
plant. Both species used as substitute for one another and have similar effects.
Calotropis is a milky latex-producing, inflexible, big, tall, much branched, perennial
shrub or small tree that grows to 5.4 m in height. They are pollinated by insects.
Calotropis gigantean can be propagated by seed or stem cutting. Seeds are dis-
persed by water and wind [32]. There are two common species of Calotropis
reported in the literature, viz., C. procera (Ait.) R.Br. and Calotropis gigantean (L.)
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1473
R.Br. mentioned by the ancient writers. Both the species consist of similar types of
phytoconstituents discovered till now and may be used as substitutes for one another
that might have similar effects [33]. Calotropis Procera is drought-resistant plant. It
quickly becomes established as a weed along degraded roadsides, waste land and in
overgrazed native pastures [34].
Papaver somniferum L.
1.9
1.9.1 Detoxification/Rectification Methods
The opium is dissolved in in rose water and filtered. The filtrate is heated till it
becomes thick for making the pills [5, 9, 10, 37]. Opium powder is heated in copper
container/pot till it becomes soft and reddish but not burned. After that hot powder
of opium is dissolved in water [10].
Aconitum Spp.
1.10
1.10.1 Detoxification/Rectification Methods
Beesh 30 g is chopped into little pieces, knotted in a fine fabric bag and dipped in a
milk-filled pot before being heated. After that, pieces of Beesh are removed and
thoroughly cleaned in water to obtain Mudabbar Beesh [5, 9 10].
Cannabis sativa L.
1.11
Cannabis sativa L. (Family: Cannabaceae) is one of the most widely used plants for
both recreational and medicinal purposes. A total of 567 natural constituents have
been discovered so far [40]. Cannabis is a dicotyledonous, angiosperm, annual
plant, normally dioecious, with male and female flowers on different plants, but can
also be monoecious, with flowers of both sexes on the same plant [41]. Commonly,
cannabis is pondered to be monospecific (Cannabis sativa L.) which is separated
into many subspecies, i.e. C. sativa subsp. sativa, C. sativa subsp. indica, C. sativa
subs. p. ruderalis, C. sativa subsp. spontanea, C. sativa subsp. afiristanca. Cannabis
is a dioecious, annual, flowering herb. Male (Staminate) plants are usually taller but
less strong than female (pistillate) plants. The stems are erect and reach a height of
0.2–2.0 m. However, mostly plants reach heights of 1–3 m [42].
Nicotiana tabacum L.
1.12
Nicotiana tabacum and Nicotiana rustica are native plants of the Americas having
evolved in the Andes around Peru/Ecuador [43]. Nicotiana tabacum L. is a tall
perennial herbaceous plant native to tropical and subtropical America that is cur-
rently grown as a cash crop all over the world. The officinal usage of N. tabacum has
been extensively reviewed elsewhere. In Ethiopian folkloric medicine, N. tabacum
is widely used for managing both human and veterinary ailments such as cancer,
ulcer, cough, snake bite and respiratory tract infections [44]. Tobacco is made from
the dried and processed leaves of the Nicotiana tabaccum plant, which is widely
planted and commercially grown in many countries. Smoking, chewing, snuffing
and dipping tobacco are the most common methods of consumption. It is most com-
monly used as a drug, and is a valuable cash crop for countries such as Cuba, India,
China and the United States [45]. With an LD50 of approximately 30–60 mg of nico-
tine in adults, death by paralysis of respiratory muscles and/or central respiratory
failure is possible. The LD50 in youngsters is approximately 10 mg [46].
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1475
2 Conclusion
References
1. Mounanga MB, Mewono L, Angone SA (2015) Toxicity studies of medicinal plants used in sub-
Saharan Africa. J Ethnopharmacol 174:618–627. https://doi.org/10.1016/j.jep.2015.06.005
2. Salgar SD, Usman RMD, Vadnere GP, Lodhi S, Patil KD (2018) Contribution of poi-
sonous plants in herbal remedies. J Pharm Bio Sci 6:18–35. https://doi.org/10.31555/
jpbs/2018/6/2/18-35
3. Soulaidopoulos S, Sinakos E, Dimopoulou D, Vettas C, Cholongitas E, Garyfallos A (2017)
Anticholinergic syndrome induced by toxic plants. World J Emerg Med 8:297–301
4. Njugi W (2018) A constructive approach on lethal plants for medicinal use. J Med Toxicol Clin
Forensic Med 4(1):1–5. https://doi.org/10.21767/2471-9641.100032
5. Rahmana S, Jahana N, Makbul SAA, Ahmad M, Gania MA (2020) Scientific appraisal of
Unani concept of islah-e-advia (rectification/purification of drugs) and its importance. J
Ethnopharmacol 258:112880. https://doi.org/10.1016/j.jep.2020.112880
6. Kalam MA, Yaqoob A, Majeed B, Ahmad A (2020) Azaraqi (Strychnos Nuxvomica L.): a novel
drug of Unani system of medicine for the management of nerve and phlegmatic diseases.
WJPLS 6:100–106
7. Lu L, Huang R, Wu Y, Jin JM, Chen HZ, Zhang LJ, Luan X (2020) Brucine: a review of phy-
tochemistry, pharmacology, and toxicology. Front Pharmacol 11:377. https://doi.org/10.3389/
fphar.2020.00377
8. Guo R, Wang T, Zhou G, Xu M, Yu X, Zhang X, Sui F, Li C, Tang L, Wang Z (2018) Botany,
phytochemistry, pharmacology and toxicity of Strychnos nux-vomica L.: a review. The. Am J
Chin Med 46:1–23. https://doi.org/10.1142/S0192415X18500015
1476 S. A. A. Makbul and S. Rahman
9. Anonymous (2007) The Unani Pharmacopoea of India. Part-1st –1st CCRUM, Ministry of
Health and Family welfare. Govt of India, New Delhi
10. Ghani N (2011) Khazainul Advia (Urdu). Idara Kitabul Shifa, New Delhi, pp 1–1375
11. Khan MA (2012) Muheete Aazm (Urdu) 1st CCRUM. Ministry of Health and Family Welfare
Govt. of India, New Delhi
12. Kabeeruddin M (2007) Makhzanul Mufradat (Urdu). Idara Kitabus Shifa, New Delhi, pp 1–44
13. Alizadeh A, Moshiri M, Alizadeh J, Balali-Mood M (2014) Black henbane and its toxicity – a
descriptive review. Avicenna J Phytomed 4:297–311
14. Al-Snafi AE (2016) Pharmacology and toxicology of Conium Maculatum- a review. Pharma
Chem J 3:136–142
15. Bhakta S, Das SK (2020) The medicinal values of Abrus precatorius: a review study. J Adv
Biotechnol Exp Ther 3:84–91. https://doi.org/10.5455/jabet.2020.d111
16. Tabasum S, Khare S, Jain K (2018) Establishment of quality standards of Abrus precatorius
Linn. Seed. Indian J Pharm Sci 80:541–546
17. Das A, Jain V, Mishra A (2016) A brief review on a traditional herb: abrus precatorius (L.). Int
J Forensic Med Toxicol Sci 1:1–10
18. Khare CP (2007) Indian medicinal plants. Springer, New Delhi
19. Anonymous (2007) The Unani Pharmacopoea of India. Part-1st –4th CCRUM, Ministry of
Health and Family welfare. Govt of India, New Delhi
20. Akbar S (2020) Introduction. In: Handbook of 200 medicinal plants. Springer, Cham. https://
doi.org/10.1007/978-3-030-16807-0_1
21. Jena J, Gupta AK (2012) Ricinus communis Linn: a phytopharmacological review. Int J Pharm
Pharm Sci 4:25–29
22. Abdul WM, Hajrah NH, Sabir JSM, Al-Garni SM, Sabir MJ, Kabli SA, Saini KS, Bora
RS (2018) Therapeutic role of Ricinus communis L. and its bioactive compounds
in disease prevention and treatment. Asian Pac J Trop Med 11:177–185. https://doi.
org/10.4103/1995-7645.228431
23. Soni P, Siddiqui AA, Dwivedi J, Soni V (2012) Pharmacological properties of Datura stramo-
nium L. as a potential medicinal tree: an overview. Asian Pac J Trop Biomed 2:1002–1008.
https://doi.org/10.1016/S2221-1691(13)60014-3
24. Trancă SD, Szabo R, Cociş M (2017) Acute poisoning due to ingestion of Datura stramo-
nium– a case report. Roman J Anaesth Intensive Care 24:65–68. https://doi.org/10.21454/
rjaic.7518.241.szb
25. Dey T, Saha S, Adhikari S, Ghosh PD (2015) A comprehensive review on medicinally impor-
tant plant, Croton tiglium L. Int J Curr Res Biosci Plant Biol 2:124–128
26. Haak M, Vinke S, Keller W, Droste J, Rückert C, Kalinowski J, Pucker B (2018) High quality
de novo transcriptome assembly of Croton tiglium. Front Mol Biosci 5. https://doi.org/10.3389/
fmolb.2018.00062
27. Liu L, Yu H, Wu H, Yang X, Pan Y, Chen Y, Wang K, Wang W, Zhang W, Jin Y, Zhang C, Jiang
A, Xia C (2017) Toxic proteins from Croton Tiglium L. exert a proinflammatory effect by
inducing release of proinflammatory cytokine and activating the P38-Mapk signaling pathway.
Mol Med Rep 16:631–638. https://doi.org/10.3892/Mmr.2017.6617
28. Anonymous (2007) The Unani Pharmacopoea of India. Part-1st –5th, CCRUM, Ministry of
Health and family welfare. Govt of India, New Delhi
29. Kabeeruddin HM (1938) Bayaze Kabeer, (Urdu). In: Hikmat book depot, vol II. Deccan,
Hyderabad, pp 1–160
30. Kumar MS, Prashant T, Kumar SP (2017) Pharmacology, phytochemistry and toxicology of
Semecarpus anacardium. Int J Pharm Sci Rev Res 42. Article No. 06:25–31
31. Gouthaman T, Kavitha MS, Ahmed BA, Kumar TS, Rao MV (2008) A Review on Semecarpus
anacardium L.: an anticancer medicinal plant. Recent progress in medicinal plant (RPMP).
Phytopharmacol Thera Values I 19:193–222
32. Merzaia AB, Riaz H, Rehman R, Nisar S, Azeem MW (2017) A review of toxicity, therapeutic
and biological activities of Calotropis. IJCBS 11:58–64
Commonly Used Poisonous Medicinal Plants in Unani System of Medicine 1477
33. Parihar G, Balekar N (2016) Calotropis procera: a phytochemical and pharmacological review.
TJPS 40:115–131
34. Verma S (2016) Calotropis Procera (Asclepiadaceae): a review. IJSRST 2:487–490
35. Masihuddin JMA, Siddiqui A, Chaudhary S (2018) Traditional uses, phytochemistry and phar-
macological activities of Papaver somniferum with special reference of Unani medicine: an
updated review. J Drug Delivery Therapeutics 8(5-s):110–1164. https://doi.org/10.22270/jddt.
v8i5-s.2069
36. Haber I, Pergolizzi J Jr, LeQuang JA (2019) Poppy seed tea: a short review and case study. Pain
Ther 8:151–155. https://doi.org/10.1007/s40122-019-0113-5
37. Nadkarni KM (2009) Indian Materia Medica 1, 3rd edn. Popular Prakashan Private
Limited, Mumbai
38. Nyirimigaboa E, Xu Y, Li Y, Wang Y, Agyemang K, Zhang Y (2015) A review on phytochem-
istry, pharmacology and toxicology studies of Aconitum. J Pharm Pharmacol 67:1–19. https://
doi.org/10.1111/jphp.12310
39. Singh MK, Vinod M, Iyer SK, Khare G, Sharwan G, Larokar YK (2012) Aconite: a pharmaco-
logical update. Int J Res Pharm Sci 3:242–246
40. Elhendawy MA, Wanas AS, Radwan MM, Azzaz NA, Toson ES, El-Sohly MA (2018)
Chemical and biological studies of Cannabis sativa roots. Med Cannabis Cannabinoids
1:104–111. https://doi.org/10.1159/000495582
41. Borille BT, Gonzáleza M, Steffensa L, Ortizb RS, Limbergera RP (2017) Cannabis sativa: a
systematic review of plant analysis. Drug Anal Res 01:1–23
42. Rajput R, Kumar R (2018) A review on Cannabis sativa: its compounds and their effects. Int
J Pharm Sci Rev Res 53:59–63
43. Musk AW, Klerk DE, NH. (2003) History of tobacco and health. Respirology 8:286–290
44. Ameya G, Manilal A, Merdekios B (2017) In vitro antibacterial activity and phytochemical
analysis of Nicotiana tabacum L. extracted in different organic solvents. Open Microbiol J
11:352–359. https://doi.org/10.2174/1874285801711010352
45. Shekins OO, Dorathy EU, Labaran ML, Joel P (2016) Phytochemical Screening of Tobacco
(Nicotiana tabacum) and Its Effects on Some Haematological Parameters and Histopathology
of Liver and Brain in Male Rats. IJBCRR 14:1–9. Article no.IJBCRR.29645. https://doi.
org/10.9734/IJBCRR/2016/29645
46. Mishra A, Chaturvedi P, Datta S, Sinukumar S, Joshi P, Garg A (2015) Harmful effects of
nicotine. Indian J Med Paediatr Oncol 36:24–31. https://doi.org/10.4103/0971-5851.151771
47. Sah A, Khatik GL, Vyas M, Yadav P (2016) A short review on anticancer investigations of
Strychnos nux-vomica. Int J Green Pharm 10:S87–S90
48. Sreedevi B, Kuchana V, Shobharani S (2021) Ethanobotanical, phytochemical and pharmaco-
logical review on Strychnos nuxvomica. J Natural Products Plant Res 11(1):1–11
49. Azhar M, Mustehasan. (2020) Phytopharmacology of an important Unani drug Bazr-Ul-Banj
(Hyoscyamus niger Linn.) – review. Asian J Pharm Clin Res 13:28–32
50. Kalam MA, Rifat I (2020) Datura species (Dhatura Safed and Dhatura Seyah): a review with
special emphasis on single-use and compound formulations and pharmacological studies rel-
evant to Unani System of Medicine. Indian J Integr Med 2:1–9
51. Sharma M, Dhaliwal I, Rana K, Delta AK, Kaushik P (2021) Phytochemistry, pharmacology,
and toxicology of datura species—a review. Antioxidants 10:1291. https://doi.org/10.3390/
antiox10081291
52. Sinsinwar S, Paramasivam I, Muthuraman MS (2016) An overview of the biological and
chemical perspectives of Croton tiglium. Pharm Lett 8:324–328
53. Gao X, Hu J, Zhang X, Zuo Y, Wang Y, Zhu S (2020) Research progress of aconitine toxicity
and forensic analysis of aconitine poisoning. Forensic Sci Res 5:25–31. https://doi.org/10.108
0/20961790.2018.1452346
54. Rawat A, Mali RR (2013) Phytochemical properties and pharmcological activities of Nicotiana
Tabacum: a review. Indian J Pharm Biol Res 1(2):74–82
55. Anonymous (1998) Qarabadeen-e-Majeedi. NCPUL Publications, pp 1–415
1478 S. A. A. Makbul and S. Rahman
56. Hkm Najm-ul Ghani (2010) Qarabadeen Najmul Gani. CCRUM, New Delhi
57. Al-Snafi AE (2018) Therapeutic importance of Hyoscyamus species grown in Iraq
(Hyoscyamus albus, Hyoscyamus Niger and Hyoscyamus reticulates)- a review. IOSR J Pharm
(IOSRPHR) 8:18–32
58. Meena PP, Chendraya PP, Praveen KM, Soundarrajan S, Srinivasan M, Sampathkumar R
(2015) Pharmacological activities of Abrus precatorius (L.) seeds. Int J Pharm Med Res
3:195–200
59. Harshavardhan B, Ashwin KSB, Ravishankar CA, Ravi M (2015) An experimental study to
assess toxic potential of jayapala (croton tiglium) beeja in relation to gastrointestinal tract. Int
J Health Sci Res 5:346–351
60. Bridgeman MB, Abazia DT (2017) Medicinal cannabis: history. Pharmacol Impl Acute Care
Setting 42:180–188
Herbal Drugs: Safety, Cost-Effectiveness,
Regulation, Current Trends, and Future
Directions
Traditional herbal medicine is the sum total of “knowledge, skill, and practices
based on the theories, beliefs, and experiences indigenous to different cultures, used
in the maintenance of health as well as in the prevention, diagnosis, improvement or
treatment of physical and mental illness” [7]. Herbal medicine has different systems
throughout the world, and all have distinctive philosophy and practices profoundly
influenced by the prevailing conditions, environment as well as geographic region
in which it first evolved [8]. However, a common philosophy is a holistic approach
(an expression of wholeness) to life, considering their emotional, physical, and
social well-being, and a focus on health rather than illness. In general, emphasis is
on the person’s total health rather than the specific affliction or conditions from
which the patient is suffering, and the use of herbs is an integral aspect of all tradi-
tional medicine systems [9–11]. Traditional Chinese medicine (TCM), date back at
least 3000 years, is a good example of how traditional knowledge can be practiced
in a holistic way in modern healthcare system [12]. Ginkgo, garlic, and ginseng,
three of the most popular botanical items, can be traced back to TCM roots and are
now used to treat various ailments [12, 13].
Herbal medicines are valued for a number of reasons all over the map. Herbal
medicines, traditional therapy, and traditional healers are the main, and often only,
source of essential healthcare for thousands of people. It is treatment that is easily
accessible to greater public, more affordable, and closer to people’s homes com-
pared with pharmaceutical drug. It is also accepted and trusted by a huge number of
individuals on a cultural level. In a time of rising healthcare expenses and practi-
cally universal austerity, the affordability of most traditional medicines makes them
all the more appealing. Traditional herbal medicine also stands up as a viable option
for dealing with the onslaught of chronic non-communicable diseases [14].
Furthermore, it is more closely aligned with the patient’s ethos, allays concerns
about the adverse reactions of pharmaceutical treatments, and meets a demand for
even more customized healthcare. Herbal medicines are primarily used to promote
public health and treatment of chronic, rather than life-threatening conditions [15–
18]. In recent decades, the disease spectrum has evolved, with complicated chronic
disorders taking center stage and results of Western medicine treatment are insuffi-
cient [19]. So, traditional medicines, particularly herbal therapy, become more prev-
alent when Western medicine fails to treat an illness, such as advanced cancer or
novel infectious diseases. Recently, herbs are used to treat a variety of chronic and
acute conditions, as well as a number of diseases and problems, including heart
disease, prostate problems, anxiety, arthritis, and to boost the immune system, to
mention a few [20, 21].
As long before recorded history, plants were used as medicines. Numerous
herbal medicines would not exist today if people had not identified the efficacy of
chemicals found in crude plant extracts when they first surfaced in the 1800s [22].
Plants are thought to be responsible for around 25% of all medications prescribed
worldwide, with 121 active chemicals in use. Exclusively 11% of the total 252 med-
ications on the World Health Organization (WHO’s) essential medicine list are
plants-derived [23]. Farnsworth and companions (1985) listed 119 chemical com-
pounds derived from plants [22], of which 88 (74%) were found as a product of
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1481
extracting the active chemicals responsible for the plant‘s traditional therapeutic
characteristics. Metformin (anti-diabetes), paclitaxel (anti-breast cancer), aspirin
(for headaches), morphine (for severe pain), digoxin (against fatal heart symptoms),
and lovastatin (to control high blood cholesterol) are only a few examples of medi-
cines produced from plants. In truth, lovastatin is derived from red yeast rice and
first documented in China during the Tang dynasty (800 AD), where it is named as
Hong Qu (Burke, 2015) [24]. These herbal medicines serve an essential role in the
treatment of several diseases and will continue to confront new challenges in the
future [25, 26].
Despite the fact that herbs are essentially of historic interest, thousands of medic-
inal herbs are to hand over-the-counter and are often used by patients all over the
world. Practitioners may find it easier to advise patients on the use of herbal medi-
cine if they have a better grasp of their authentication as well as regulation, safety,
and efficacy [6].
The phrase “High Value Minor Crops” refers to plants that are relatively small con-
tributors to a country’s agricultural output. Despite being little contributions to out-
put, the significance of these plants in aggregate can be shown in the fact that their
global trade in 2006 generated US$ 60 billion [27, 28]. Annual herbal medicines
expenditures in the Republic of Korea increased from US$4.4 billion in 2004 to
US$7.4 billion in 2009 [29]. The annual “out of pocket” expenses for natural prod-
uct in the United States was $14.8 billion in 2008 [30]. Americans spent $33.9 bil-
lion on herbal medicine in 2009, up 25% from the $27 billion they spent on the same
treatments in 1997 [31]. In certain nations, such as Singapore and the Republic of
Korea, where the traditional healthcare system is well-established, herbal medicine
is still widely used by 76 and 86% of the people, respectively [29]. In 2012, the
output of Chinese materia medica was estimated to be worth US$83.1 billion, up
more than 20% from the previous year [5]. Pakistan’s high-value plant exports
reached over US$10.5 million in 2012, with Swat District accounting for a large
portion of the supply [32]. Now the global market of herbal medicines is about 100
billion (see http://www.nutraceuticalsworld.com/). Because of the variety of legis-
lation and regulatory categories for traditional herbal medicine, estimating the size
of the market for traditional herbal medicines across member states is difficult.
However, available evidence indicates that there is a substantial market.
3 Regulation
Herbs are classified as dietary supplements under the “Dietary Supplement Health
and Education Act (DSHEA) of 1994 [33].” Under this law, supplements are defined
as “anything that supplements the diet.” Vitamins, minerals, herbs, amino acids,
1482 S. N. Ahmed et al.
enzymes, organ tissues, metabolites, extracts, and concentrations are all examples
of supplements. Dietary supplements cannot claim to “diagnose, cure, mitigate,
treat, or prevent illness,” which is a fundamental distinction between drugs and
supplements [33]. It’s worth mentioning that dietary supplement companies are
authorized to make certain “structure/function” claims, which are generally vague
health benefits claims. For example, an Echinacea product can promise to “boost the
body’s natural defences” (it’s widely used to cure or prevent the common cold) [6].
As herbs are defined as dietary supplements under the current law, hence manu-
facturers can sell and market their herbal medicines without demonstrating safety
and efficacy as is mandatory for pharmaceutical drugs. Also, unlike medicines,
where a company must provide the Food and Drug Administration (FDA) with evi-
dence of safety and efficacy before a product can be sold, the FDA faces the regula-
tory burden of establishing that a dietary supplement is harmful before it can be
removed from the market [33]. This regulatory framework, predictably, has resulted
in issues with herbal medicine consistency and safety. Many recent studies have
reported that the quantities of probable active components in herbal preparations
vary substantially. For example, a recent study of 25 ginseng products indicated a
variation of 15- to 36-fold and 43- to 200-fold in the concentration of two biologi-
cally active ingredients: ginsenosides and eleutherosides, respectively [34]. As a
result, it may be problematic for patients to determine the precise composition of
the drugs they are interested in taking.
However, WHO has perceived that herbal medicine makes a significant contribu-
tion to providing vital healthcare [35]. The Office of Alternative Medicine was
established within the National Institutes of Health in 1989 to promote scientific
studies in the field of herbal medicine (http://nccam.nih.gov), and the European
Scientific Cooperative on Phytotherapy (ESCOP) was established in 1989 with the
intention of promoting the scientific status and harmonization of herbal medicines
at the European level (https://escop.com/). As a result, investment in the evaluation
of herbal treatments has increased. While this investment is little in comparison to
the pharmaceutical industry’s total investment in research and development, it nev-
ertheless shows genuine public, enterprise, and government concern in this field [36].
With the great growth in interest in and use of traditional medicines around the
world, two major areas of interest have emerged, each posing significant concerns.
These are a wide range of worldwide and national policies governing the production
and use of herbs (and other complementary medicines), as well as their authenticity,
quality, safety, and scientific evidence regarding health claims [8, 37].
A national survey in United States documented that the use of an herb to treat a
medical illness within the past year was 18.9% when meal supplements excluded
[38]. Bent (2008) reviewed the evidence for efficacy of most commonly used herbs
(including Echinacea, Ginseng, Ginkgo, Garlic, St. John’s wort, Peppermint,
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1483
Ginger, Soy, Chamomile and Kava), reported that systemic reviews supported effi-
cacy for five out of ten common herbs (Garlic, Ginkgo, St. John’s wort, Soy, and
Kava) for specific indications Table 1 [6] and discussed below.
Garlic has been extensively researched due to several claims of medicinal prop-
erties. But the most substantial body of research documented the effect on choles-
terol and hypertension [40, 41]. For lowering cholesterol, the most recent systematic
review identified 10 randomized, placebo-controlled trials; six of ten studies found
garlic to be effective. The average drop in total cholesterol was about 9.9–11.4%
[42]. The existing evidence suggests that garlic is superior to placebo in lowering
cholesterol levels [40]. However, the magnitude of the effect is modest in compared
to the 17–32% reduction observed with active FDA-approved drug “statin” medica-
tions [43]. Garlic against hypertension has been studied in seven randomized,
placebo-controlled trials. This meta-analysis found that garlic had a significant
decreasing effect on both systolic weighted mean difference (WMD: 6.71 mmHg;
95% confidence interval [CI]: 12.44 to 0.99; P = 0.02) and diastolic blood pressure
(BP; WMD: 4.79 mmHg; 95% CI: 6.60 to 2.99; P 0.00001) when compared to the
placebo. There were no major side effects noted [41]. It is, however, associated with
gastrointestinal pain and poor garlic breath [40]. It’s also been linked to a higher risk
of blood thinning after using garlic [44, 45].
Ginkgo extracts are one of the well-studied herbal formulation, with flavonoids
and terpenoids standardized to 24% and 6%, respectively. Despite mixed results in
previous trials, ginkgo is generally effective for mild dementia, with a small ben-
efit of about 3% in the Alzheimer’s Disease Assessment Scale-Cognitive subtest [46].
It is important to mention that ginkgo was not effective for improving cognitive
function in elderly patients without dementia [47]. In addition, ginkgo was reported
to enhance pain-free walking distance in claudication patients in a systematic review
of eight trials, a minor benefit with dubious clinical value [40]. While the side
effects of ginkgo and placebo were similar in clinical trials, [48] the documented
link between ginkgo and spontaneous bleeding is a major issue [49].
Herbal medicine has long been practiced for disease treatment and prevention, as
well as for health promotion and prolonging one’s life span and quality of life.
However, a systematic approach to evaluating their safety and effectiveness is lack-
ing. Many individuals find herbal therapy appealing because of its holistic approach
to health treatment, but scientific evaluation is difficult because so many aspects
must be considered. Herbal medicines are widely used and often perceived as “natu-
ral” and therefore safe. They are commonly used in combination with other medica-
tions and are derived from plants, which vary in species, growth conditions, and
biologically active ingredients. Herbal extracts can be contaminated, adulterated, or
contain harmful substances [64, 65]. There are many case reports of nephropathy
induced by the use of some Chinese herbal medicines. A particularly morbid case
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1485
series involves 105 patients in Belgium who suffered nephropathy after consuming
a Chinese herbal medicine (Aristolochia fangchi) for weight loss. End-stage renal
failure affected 43 people, and 39 had their kidneys removed as a precaution.
Urothelial cancer was detected in 18 of these patients, which was linked to the syn-
thesis of DNA adducts from the aristolochic acid in this medicinal herb [66].
Pyrrolizidine alkaloids, which are complex compounds found in certain plants and
may be intentionally or mistakenly added to herbal remedies, are another major
source of toxicity in herbal medicines (for example; comfrey, which is still available
in the United States). Certain alkaloids cause hepatotoxicity by inducing sinusoidal
obstruction syndrome (SOS) that can rapidly progressive and be lethal [67].
Herbal medicine quality management has a direct impact on their safety and
efficacy [64, 65]. However, due to a lack of suitable policies or regulatory require-
ments, as well as a lack of adequate or approved research technique for evaluating
traditional medicines, there is little data on the composition and quality of most
herbal medicines [68, 69]. Furthermore, because the drug approval process does not
allow for undifferentiated mixes of natural compounds, there is relatively little
research on entire herbal mixtures. Isolating each active chemical from each herb
would be extremely time-consuming and expensive, rendering it uneconomical for
manufacturers [70]. Another concern is that, despite the increasing demand of
botanical dietary and herbal supplements, some herbal items on the market are
likely to be of poor quality and efficacy, even though the herbal product has been
proven to have an effect in controlled research using high-quality sources. Herbs, as
natural products, are believed to be intrinsically harmless with no adverse effects,
and that efficacy can be attained at a wide range of doses. However, there are no
established “doses” for herbs, and different drug interactions and undesirable side
effects are possible.
The presence of numerous active components that collectively give a potentiat-
ing effect, may not be achieved by any single active compound, is a key prospective
advantage of botanicals over mainstream single-component medications. This
advantage poses a challenge for the isolation and identification of bioactive con-
stituents. To validate in vivo efficacy, products that are identified by activity-guided
fractionation must be tested in approved model organisms. Preferably, plants should
be grown under controlled conditions particularly when used as a source of botani-
cal extracts and originate from a characterized and uniform genetic source with a
taxonomic record of the genus, species, and cultivar or other extra signifier, and the
concentration of the whole plant extract should be standardized and free of any pos-
sible hazards. The source of the seed, cultivation conditions, location, and any
chemical treatment such as pesticides should all be recorded. Plant extracts can vary
from year to year and may be considerably affected by temperature, drought, or
flood, and geographic location, because the environment can have a considerable
impact on phytochemical profiles and the efficacy of the plant end product. For that
reason, biochemical profiling must be applied to confirm that the product is made
from consistent material. The process of concentrating the bioactive chemicals can
be difficult and challenging and might have a negative impact on their solubility and
bioavailability.
1486 S. N. Ahmed et al.
Over the previous two decades, there has been an expanding body of scientific
knowledge on the efficacy of herbal medicine, understanding of underlying mecha-
nisms, and progress in methods to ensure quality and standard of materials and
products. Research partnerships are primarily focusing on high burden of disease
areas where conventional medicine has produced mixed results, especially in the
prevention and treatment of chronic disease, and towards improving outcomes by
combining complementary and conventional interventions. Once efficacy and safety
have been demonstrated, analyzing the cost-effectiveness of medical interventions,
whether conventional or alternative, is crucial for patients, practitioners, and
policymakers.
Cost-effectiveness analysis (CEA) is a type of economic analysis that analyzes
the relative expenses (costs) and benefits (effects) of several courses of action. CEA
differs from cost-benefit analysis in that it puts a monetary value to the measures of
effect [72].
In past few years, the range of economic evaluations of herbal medicine has
improved [73], with many herbal therapies being demonstrated to be cost-effective
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1487
when compared to standard care. In order to account for the entire benefit of herbal
therapies, economic evaluation of herbal medicine logically should incorporate
proper process and outcome-based evaluation of the humanistic perspective that
characterize its philosophical doctrinism [74].
The unavailability of administrative claims makes it challenging to collect eco-
nomic outcome data because many herbal therapies are accessible over-the-counter
and/or are frequently paid for out-of-pocket. Furthermore, many over-the-counter
drugs, such as ST JOHN’S WORT, are unstandardized and of variable quality. Both
the therapy’s costs and outcomes will be influenced by standardization and quality.
Even if the clinical outcomes of a herbal therapy are equivalent to or somewhat less
favorable than those of conventional medicine, the reduced cost of healthcare can
make such therapies more appealing to decision makers [75].
In many meta-analyses of randomized controlled trials, Hypericum perforatum
formulations were found to be superior to placebo [76, 77] and equally effective as
standard antidepressants in the acute treatment of mild to moderate depression [78].
This benefit has the potential to influence how prescribed medicines, such as antide-
pressants, are used.
Numerous treatment parameters have the ability to influence the treatment’s
cost-effectiveness. In the case of St John’s wort, a distinct benefit over antidepres-
sants has been documented in terms of decreased treatment withdrawal rates, low
rates of side effects, and strong compliance, all of which are important factors in
determining the cost-effectiveness of a certain form of therapy [79].
Therefore, any evidence of therapeutic benefit and likely superiority of St John’s
wort over standard therapies could interpret into obvious cost-effectiveness.
The most significant risk associated with use is the possibility of drug interac-
tions, which can be reduced by using preparations with modest hyperforin levels
[80]. The relatively low cost of Hypericum perforatum extract makes it worth con-
sidering in the cost-effectiveness analysis of mild to moderate depression treat-
ments, given that the indirect costs of depression are greater than five times direct
treatment costs, and given the rising cost of standard antidepressants [75].
Phytodolor™, a patented herbal medicine, when compared to the main non-
steroidal anti-inflammatory drug Diclofenac, was found to be less expensive in the
treatment of osteoarthritis. The literature review presented similarity of efficacy and
clinical outcomes, with cost thus being the most important factor of cost-
effectiveness [81].
Furthermore, numerous meta-analyses have shown that acupuncture could be a
credible prophylactic treatment option for migraineurs who suffer from frequent or
uncontrolled migraines, as well as those who are experiencing drug side effects [82,
83]. In terms of incidence of reduced side effects and potential long-term efficacy
after quitting, acupuncture appears to be superior than mainstream treatment [84–
86]. There has been some research that introducing acupuncture to an existing
mainstream treatment can decrease costs by minimizing use of prescription drug,
rate of sick absence, and medical assessment appointments [87]. Despite the over-
whelming evidence for acupuncture’s potential cost-effectiveness in chronic pain
1488 S. N. Ahmed et al.
management, like headache, osteoarthritis, and lower back pain, more research on
acupuncture’s long-term cost-effectiveness is needed [86–89].
A review of the medical, corporate, and payer literature reveals that, to start,
immediate and significant health benefits and cost savings could be realized through-
out our healthcare system by utilizing three integrative strategies: (1) integrative
lifestyle change programs for those with chronic disease, (2) integrative interven-
tions for people experiencing depression, and (3) integrative preventive strategies to
support wellness in all populations [90].
Research demands in the field of herbal medicines are tremendous, but they are bal-
anced by the potential health advantages and enormous size of global herbal medi-
cine market. Expended research on the authenticity, safety, pharmacological effects,
and clinical efficacy of the many herbs in regular use is much needed [21, 91, 92].
Several experts have already proposed a number of significant reforms to the
regulation of herbal products that could boost their quality and adequate use [93,
94]. These include requiring producers to register with the FDA, implementing
safety checks just like to those necessary for over-the-counter medicines, requiring
any health claims to be backed up by FDA-approved research, and ensuring that
product labels include a concise list of all contents. While these measures will
undoubtedly improve herbal product safety, more adjustments are required to
improve and promote high-quality research. To maintain uniformity amongst
research, the most important aspect will be to develop definite standards for herbal
medicines [95]. For definitive authenticity and quality of herbal medicines, genomic
testing and chemical fingerprinting procedures using hyphenated techniques are
now available. They should be regulated to safeguard the consumers, but uncertain-
ties about efficacy will continue until sufficient amounts of scientific evidence from
experimental and controlled clinical studies accumulate [21, 91, 92]. Since no evi-
dent, well-established criterion of the chemical fingerprint exists for the most widely
used herbs, so recent researches (such as the aforementioned study of Echinacea)
[96] that even adopted high-quality methodology have always been open to criti-
cism about the composition of the herbal medicine used [95]. Generally, interna-
tional studies on herbal medicines should be conducted to certain ethical standards
as all other research associated with human subjects, with data shared among coun-
tries. Collaboration, cultural value, scientific validity, fair subject selection, favor-
able risk-benefit ratio, independent review, informed consent, and respect for the
subjects should all be considered [21, 91]. However, conducting large, controlled
human trials on the clinical effectiveness of herb is cost prohibitive, especially if the
goal is to promote human well-being. Therefore, innovative biomarkers that are
more clearly inked to health (and illness) outcomes are urgently needed. It is neces-
sary to develop predictive biomarkers and subtle but detectable signs of early
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1489
cellular and molecular changes that can be linked to the emergence of specific dis-
eases [97].
There should be research-based information on whether whole herbs or extracted
chemicals are better to meet the problems of finding the active compounds in the
plants. The subject of drug interactions is also of a great value that demands pro-
gressive awareness and studies, as polypharmacy and polyherbacy [15–18].
If an adequately funded branch of the FDA supports and coordinates the effort to
set standards for herbal medicines, it is most likely to succeed. Research would also
pursue more promptly if a few type of patent protection is available, so that the
producers that invest in clinical trials and document the safety and efficacy of their
products could be remunerated financially. Finally, techniques to improve clinical
research progress would assist in addressing the “backlog” of 20,000 herbs with
insufficient proof of safety and efficacy [95]. Although this technique is still being
investigated, it has the ability to dramatically shorten the time and cost of clinical
trials of herbal medicines.
8 Conclusion
References
1. Dar RA, Shahnawaz M, Qazi PH (2017) General overview of medicinal plants: a review. J
Phytopharm 6(6):349–351
2. Joharchi MR, Amiri MS (2012) Taxonomic evaluation of misidentification of crude herbal
drugs marketed in Iran. Avicenna J Phytomed 2(2):105
3. Swerdlow J (2000) Modern science embraces medicinal plants. In: Nature’s medicine: plants
that heal. National Geographic Society, Washington, DC, pp 110–157
4. World Health Organization (2019) WHO global report on traditional and complementary med-
icine 2019. World Health Organization
5. World Health Organization (2013) WHO traditional medicine strategy: 2014–2023. World
Health Organization
6. Bent S (2008) Herbal medicine in the United States: review of efficacy, safety, and regulation.
J Gen Intern Med 23(6):854–859
7. World Health Organization. W.H.O. Traditional Medicine 2010, July 21. Available from:
http://www.who.int/topics/traditional_medicine/en/
8. World Health Organization (2005) National policy on traditional medicine and regulation of
herbal medicines: report of a WHO global survey. World Health Organization
9. Conboy L et al (2007) The relationship between social factors and attitudes toward conven-
tional and CAM practitioners. Complement Ther Clin Pract 13(3):146–157
10. Rishton GM (2008) Natural products as a robust source of new drugs and drug leads: past suc-
cesses and present day issues. Am J Cardiol 101(10):S43–S49
11. Schmidt B et al (2008) A natural history of botanical therapeutics. Metabolism 57:S3–S9
12. Xutian S, Zhang J, Louise W (2009) New exploration and understanding of traditional Chinese
medicine. Am J Chin Med 37(03):411–426
13. Li W-F, Jiang J-G, Chen J (2008) Chinese medicine and its modernization demands. Arch Med
Res 39(2):246–251
14. World Health Organization (2009) World health statistics 2009. World Health Organization
15. Canter PH, Ernst E (2004) Herbal supplement use by persons aged over 50 years in Britain.
Drugs Aging 21(9):597–605
16. Qato DM et al (2008) Use of prescription and over-the-counter medications and dietary sup-
plements among older adults in the United States. JAMA 300(24):2867–2878
17. Loya AM, González-Stuart A, Rivera JO (2009) Prevalence of polypharmacy, polyherbacy,
nutritional supplement use and potential product interactions among older adults living on the
United States-Mexico border. Drugs Aging 26(5):423–436
18. Cohen PA, Ernst E (2010) Safety of herbal supplements: a guide for cardiologists. Cardiovasc
Ther 28(4):246–253
19. Zhang J et al (2015) The safety of herbal medicine: from prejudice to evidence. Hindawi
20. De Smet PA (2005) Herbal medicine in Europe—relaxing regulatory standards. N Engl J Med
352(12):1176–1178
21. Tilburt JC, Kaptchuk TJ (2008) Herbal medicine research and global health: an ethical analy-
sis. Bull World Health Organ 86:594–599
22. Farnsworth NR et al (1985) Medicinal plants in therapy. Bull World Health Organ 63(6):965
23. Rates SMK (2001) Plants as source of drugs. Toxicon 39(5):603–613
24. Burke FM (2015) Red yeast rice for the treatment of dyslipidemia. Curr Atheroscler Rep
17(4):22
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1491
25. Ali I et al (2010) Natural products: human friendly anti-cancer medications. Egypt Pharm J
(NRC) 9(2):133–179
26. Lu D et al (2016) Discover natural chemical drugs in modern medicines. Metabolomics (Los
Angel) 6(181):2153-0769.1000181
27. Adhikari B (2001) Socio-economic heterogeneity and income distribution: evidence from
common property resource management. J Forest Livelihoods 1(1):22–24
28. Hamilton A (2013) Plant conservation: an ecosystem approach. Routledge
29. World Health Organization (2012) The regional strategy for traditional medicine in the Western
Pacific (2011–2020)
30. Barnes PM, Bloom B, Nahin RL (2008) Complementary and alternative medicine use among
adults and children; United States, 2007
31. Greg Arnold D. 2010. Study cites cost savings of omega-3 fats and St John’s Wort. http://www.
nowfoods.com/BasicArticles/080683.htm
32. Sher H et al (2014) Economic benefits of high value medicinal plants to Pakistani communi-
ties: an analysis of current practice and potential. J Ethnobiol Ethnomed 10(1):1–16
33. Shneeman B et al (2005) Review of liver-related risks for chaparral. In: Dietary supplements:
a framework for evaluating safety
34. Harkey MR et al (2001) Variability in commercial ginseng products: an analysis of 25 prepara-
tions. Am J Clin Nutr 73(6):1101–1106
35. World Health Organization. W.H.O. Traditional medicine. Available from: http://www.who.
int/topics/traditional_medicine/en/
36. Li JW-H, Vederas JC (2009) Drug discovery and natural products: end of an era or an endless
frontier? Science 325(5937):161–165
37. Sahoo N, Manchikanti P, Dey S (2010) Herbal drugs: standards and regulation. Fitoterapia
81(6):462–471
38. Barnes PM et al (2004) Complementary and alternative medicine use among adults: United
States, 2002. In: Seminars in integrative medicine. Elsevier
39. Bent S, Ko R (2004) Commonly used herbal medicines in the United States: a review. Am J
Med 116(7):478–485
40. Stevinson C, Pittler MH, Ernst E (2000) Garlic for treating hypercholesterolemia: a meta-
analysis of randomized clinical trials. Ann Intern Med 133(6):420–429
41. Xiong X et al (2015) Garlic for hypertension: a systematic review and meta-analysis of ran-
domized controlled trials. Phytomedicine 22(3):352–361
42. Alder R et al (2003) A systematic review of the effectiveness of garlic as an anti-hyperlipidemic
agent. J Am Acad Nurse Pract 15(3):120–129
43. Koren MJ (2005) Statin use in a “real-world” clinical setting: aggressive lipid lowering com-
pared with usual care in the aggressive lipid-lowering initiation abates new cardiac events
(ALLIANCE) trial. Am J Med 118(12):16–21
44. Rose KD et al (1990) Spontaneous spinal epidural hematoma with associated platelet dysfunc-
tion from excessive garlic ingestion: a case report. Neurosurgery 26(5):880–882
45. Linde K et al (1998) St John’s wort for depression. Cochrane Database Syst Rev (4)
46. Oken BS, Storzbach DM, Kaye JA (1998) The efficacy of Ginkgo biloba on cognitive function
in Alzheimer disease. Arch Neurol 55(11):1409–1415
47. Solomon PR et al (2002) Ginkgo for memory enhancement: a randomized controlled trial.
JAMA 288(7):835–840
48. Birks J, Grimley E, Van Dongen M (2002) Ginkgo biloba for cognitive impairment and demen-
tia (Cochrane review). Cochrane Libr 4:1–19
49. Bent S et al (2005) Spontaneous bleeding associated with Ginkgo biloba. J Gen Intern Med
20(7):657–661
50. Brevoort P (1998) The blooming US botanical market: a new overview
51. Carpenter C et al (2008) Hypericum and nurses: a comprehensive literature review on the effi-
cacy of St. John’s wort in the treatment of depression. J Holist Nurs 26(3):200–207
1492 S. N. Ahmed et al.
52. Linde K et al (1996) St John’s wort for depression—an overview and meta-analysis of ran-
domised clinical trials. BMJ 313(7052):253–258
53. Butterweck V (2003) Mechanism of action of St John’s wort in depression. CNS Drugs
17(8):539–562
54. Zanoli P (2004) Role of hyperforin in the pharmacological activities of St John’s wort. CNS
Drug Rev 10(3):203–218
55. Shelton RC et al (2001) Effectiveness of St John’s wort in major depression: a randomized
controlled trial. JAMA 285(15):1978–1986
56. Hammerness P et al (2003) St. John’s wort: a systematic review of adverse effects and drug
interactions for the consultation psychiatrist. Psychosomatics 44(4):271–282
57. Lethaby A et al (2007) Phytoestrogens for vasomotor menopausal symptoms. Cochrane
Database Syst Rev (4)
58. Taku K et al (2007) Soy isoflavones lower serum total and LDL cholesterol in humans: a meta-
analysis of 11 randomized controlled trials. Am J Clin Nutr 85(4):1148–1156
59. Pittler MH, Ernst E (2003) Kava extract versus placebo for treating anxiety. Cochrane Database
Syst Rev (1)
60. Control, C.f.D. and Prevention (2002) Hepatic toxicity possibly associated with kava-
containing products--United States, Germany, and Switzerland, 1999–2002. MMWR Morb
Mortal Wkly Rep 51(47):1065–1067
61. Rotblatt M, Ziment I (2002) Evidence-based herbal medicine. Hanley & Belfus
62. Ulbricht CE, Basch EM (2005) Natural standard herb & supplement reference: evidence-based
clinical reviews. Mosby
63. Fugh-Berman A (2003) The 5-minute herb and dietary supplement consult. Lippincott
Williams & Wilkins
64. Ernst E, Schmidt K, Wider B (2005) CAM research in Britain: the last 10 years. Complement
Ther Clin Pract 11(1):17–20
65. Ribnicky DM et al (2008) Evaluation of botanicals for improving human health. Am J Clin
Nutr 87(2):472S–475S
66. Nortier JL et al (2000) Urothelial carcinoma associated with the use of a Chinese herb
(Aristolochia fangchi). N Engl J Med 342(23):1686–1692
67. Stickel F, Patsenker E, Schuppan D (2005) Herbal hepatotoxicity. J Hepatol 43(5):901–910
68. World Health Organization (2000) General guidelines for methodologies on research and eval-
uation of traditional medicine. World Health Organization
69. Kantor M (2009) The role of rigorous scientific evaluation in the use and practice of comple-
mentary and alternative medicine. J Am Coll Radiol 6(4):254–262
70. Applequist W (2003) Herbal medicine. Chaos in the Marketplace. Econ Bot 57(1):150–151
71. Brower V (2008) Back to nature: extinction of medicinal plants threatens drug discovery.
Oxford University Press
72. Bleichrodt H, Quiggin J (1999) Life-cycle preferences over consumption and health: when is
cost-effectiveness analysis equivalent to cost–benefit analysis? J Health Econ 18(6):681–708
73. Herman PM, Craig BM, Caspi O (2005) Is complementary and alternative medicine (CAM)
cost-effective? A systematic review. BMC Complement Altern Med 5(1):1–15
74. Van Schaik DJ et al (2004) Patients’ preferences in the treatment of depressive disorder in
primary care. Gen Hosp Psychiatry 26(3):184–189
75. Solomon D et al (2011) Potential of St John’s wort for the treatment of depression: the eco-
nomic perspective. Australian New Zealand J Psychiatry 45(2):123–130
76. Linde K, Berner MM, Kriston L (2008) St John’s wort for major depression. Cochrane
Database Syst Rev (4)
77. Gastpar M, Singer A, Zeller K (2006) Comparative efficacy and safety of a once-daily dosage
of hypericum extract STW3-VI and citalopram in patients with moderate depression: a double-
blind, randomised, multicentre, placebo-controlled study. Pharmacopsychiatry 39(02):66–75
78. Linde K, St. (2009) John’s wort–an overview. Compl Med Res 16(3):146–155
Herbal Drugs: Safety, Cost-Effectiveness, Regulation, Current Trends, and Future… 1493
79. Sullivan PW et al (2004) A comparison of the direct costs and cost effectiveness of serotonin
reuptake inhibitors and associated adverse drug reactions. CNS Drugs 18(13):911–932
80. Whitten D et al (2006) The effect of St John’s wort extracts on CYP3A: a systematic review of
prospective clinical trials. Br J Clin Pharmacol 62(5):512–526
81. Bensoussan A (2010) Cost effectiveness of complementary medicines
82. Linde K et al (2016) Acupuncture for the prevention of episodic migraine. Cochrane Database
Syst Rev (6)
83. Coeytaux RR, Befus D (2016) Role of acupuncture in the treatment or prevention of
migraine, tension-type headache, or chronic headache disorders. Headache J Head Face Pain
56(7):1238–1240
84. Kim M-R et al (2016) Safety of acupuncture and pharmacopuncture in 80,523 musculoskeletal
disorder patients: a retrospective review of internal safety inspection and electronic medical
records. Medicine 95(18)
85. Zhang J et al (2010) Acupuncture-related adverse events: a systematic review of the Chinese
literature. Bull World Health Organ 88:915–921
86. MacPherson H et al (2017) The persistence of the effects of acupuncture after a course of treat-
ment: a meta-analysis of patients with chronic pain. Pain 158(5):784
87. Kim S-Y et al (2012) A systematic review of cost-effectiveness analyses alongside randomised
controlled trials of acupuncture. Acupunct Med 30(4):273–285
88. Ambrósio E, Bloor K, MacPherson H (2012) Costs and consequences of acupuncture as a
treatment for chronic pain: a systematic review of economic evaluations conducted alongside
randomised controlled trials. Complementary Therapies in Medicine 20(5):364–374
89. Deng Z-Q et al (2012) Health economic evaluation of acupuncture along meridians for treat-
ing migraine in China: results from a randomized controlled trial. BMC Complement Altern
Med 12(1):1–7
90. Guarneri EM, Horrigan BJ, Pechura CM (2010) The efficacy and cost effectiveness of integra-
tive medicine: a review of the medical and corporate literature. Explore 6(5):308–312
91. Giordano J, Engebretson J, Garcia MK (2005) Challenges to complementary and alternative
medical research: focal issues influencing integration into a cancer care model. Integr Cancer
Ther 4(3):210–218
92. Evans S (2008) Changing the knowledge base in Western herbal medicine. Soc Sci Med
67(12):2098–2106
93. Lewis JD, Strom BL (2002) Balancing safety of dietary supplements with the free market. Ann
Intern Med 136(8):616–618
94. Marcus DM, Grollman AP (2002) Botanical medicines—the need for new regulations. Mass
Medical Soc 347(25):2073–2076
95. Jacobs BP et al (2005) An internet-based randomized, placebo-controlled trial of kava and
valerian for anxiety and insomnia. Medicine 84(4):197–207
96. Turner RB et al (2005) An evaluation of Echinacea angustifolia in experimental rhinovirus
infections. N Engl J Med 353(4):341–348
97. Benzie IF, Wachtel-Galor S (2011) Herbal medicine: biomolecular and clinical aspects
Therapeutic Properties of Herbal
Constituents Subjected for Clinical Trials
1 Introduction
Nature is source of variety of medicinal products from the very ancient time, with
numerous varieties of drugs useful for the mankind and directly produced from the
plant sources. One particularly strong application of Penicillin’s discovery, together
with the fact that many medications come from microorganisms, etc. In the late
1980s, combinatorial chemistry diverted the focus of efforts of drug discovery
directly from nature to the bench of laboratories, which was further subjected for
clinical trials.
However, natural compounds also face several kinds of challenges in the discov-
ery of new drugs, viz., barriers of technical screening, isolation of new compounds,
and characterization and optimization of the same, which has directly contributed to
rejection in their quest by the pharmacy industries since the early 1990s till now.
Nowadays, several developments in the field of technology exist, which involves
improvement in use of analytical tools, genome studies, and various engineering-
oriented strategies, and advances in microbial culturing, etc., are opening up new
opportunities in the field of research.
New drug discovery with the use of natural products is a very demanding task in
the field of research for the designing of new compound leads. It provides descrip-
tion of the bioactive compounds originating from natural resources, analysis on the
basis of presence of phytochemicals, and their categorization and investigation on
E. Vatsa (*)
Faculty of Pharmaceutical Sciences, ICFAI University, Baddi,
Solan, Himachal Pradesh, India
M. Aggarwal
Department of Pharmacy, Himachal Institute of Pharmacy,
Paonta Sahib, Himachal Pradesh, India
the basis of therapeutic evidence, because this is the base with which any drug is
subjected for clinical trials.
This chapter provides a brief description about some common natural products
and also reviews clinical trials performed on these natural products. The possible
mechanisms of action for the practical impact of the following natural compounds
are also described.
1.1 Garlic
Garlic (Allium sativum) is an aromatic yearly herbaceous spice and one of the most
seasoned and authenticated herbs that have been used since ancient times. It belongs
to Amaryllidaceae family [1, 2]. Garlic is utilized as a remedy for numerous com-
mon maladies because of hundreds of phytochemicals present in it [2]. Reportedly
it includes sulfur-containing compounds such as ajoenes (E-ajoene, Z-ajoene), thio-
sulfinates (allicin), vinyldithiins (2-vinyl-(4H)-1,3-dithiin; 3-vinyl-(4H)-1,2-
dithiin), sulfides (diallyl disulfide [DADS], diallyl trisulfide [DATS]), and others,
which account for 82% of the total garlic sulfur content [3].
Garlic has solid antioxidant properties due to its dietary and phenolic com-
pounds. An orderly survey and meta-analysis of 12 randomized controlled trials
(RCTs) uncovered noteworthy increments in serum antioxidant capacity and super-
oxide dismutase levels and diminished serum malondialdehyde levels as a result of
garlic (Allium sativum) supplementation (80–4000 mg/day for 2–24 weeks) [2].
Garlic extract was found to increase the activities of some antioxidant enzymes
(e.g., superoxide dismutase [SOD]) and decrease glutathione peroxidase (GSH-Px)
in rat liver tissues. Notably, several reports indicated that aged garlic extract (AGE)
rich in flavonoids, phenol, and different sulfur compounds, e.g., S-ally-(L)-cysteine
(SAC), shows high radical scavenging activity [4]. Although experimental studies
have shown a clear hypoglycemic effect of garlic, the effect of garlic on human
blood glucose remains controversial. Garlic significantly lowered total cholesterol
and low-density lipoprotein (LDL) cholesterol and moderately increased high-
density lipoprotein (HDL) cholesterol compared to placebo in diabetic patients [5].
In a double-blind clinical trial (N = 38), a combination of nettle leaves (20% w/w),
onion and garlic (20%), fenugreek seeds (20%), walnut leaves (20%), cinnamon
bark (10%), and berry leaf (10%) powder effectively controlled type 2 diabetes in
human subjects [6]. Aged garlic extract (2400 mg/day orally) reduced volumes of
low attenuation but not total fibrous or fibro fatty plaque in the coronary arteries of
patients with diabetes mellitus (N = 80) compared with placebo [7]. Crushed raw
garlic (100 mg/kg twice daily for 4 weeks) reduced waist circumference, systolic
and diastolic blood pressure, triglycerides, and fasting blood glucose, and increased
serum HDL cholesterol in patients with metabolic syndrome [8]. In an open-label
phase 1 trial, a combination of A. sativum, Aloe vera, Nigella sativa, Plantago psyl-
lium, Silybum marianum, and Trigonella foenum-graecum reduced fasting blood
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1497
glucose, HbA1, LDL, and triglycerides in patients with hyperlipidemia and hyper-
glycemia with advanced type 2 diabetes [9].
Garlic and its preparations are widely known as agents for the prevention and
treatment of cardiovascular diseases. Extensive scientific literature supports the
suggestion that garlic consumption has significant effects on lowering blood pres-
sure, preventing atherosclerosis, reducing cholesterol and triglycerides, inhibiting
platelet aggregation, and increasing fibrinolytic activity [25]. In a randomized con-
trolled trial (N = 104), taking aged garlic extract (2400 mg daily for 1 year) slowed
the development of coronary artery calcification and progression to coronary ath-
erosclerosis, with a significant decrease in systolic blood pressure, in cardiovascular
disease patients [10]; also, aged garlic extract decreased the cardio-ankle vascular
index, a measure of endothelial function and arterial stiffness, over a three-month
period in subjects with type 2 diabetes mellitus (N = 65) [11]. In a randomized,
placebo-controlled study, black garlic, given for 6 months, improved heart function,
as measured by walking distance, left ventricular ejection fraction (LVEF), and
quality of life, in patients with coronary artery disease [12]. The preventive effect of
garlic on atherosclerosis has been attributed to its ability to reduce the lipid content
in the arterial membrane. Allicin, S-allyl cysteine, present in aged garlic extract and
diallyl disulfide, present in garlic oil, are the active compounds responsible for the
anti-atherosclerotic effect [13].
Many in vitro and in vivo studies are done to know the cancer-preventive effects
of garlic preparations and their respective components. It is apparent that garlic
contains a large number of potent bioactive compounds with anticancer properties,
largely derived from allyl sulfide. Different garlic derivatives have been reported to
modulate an increasing number of molecular mechanisms in carcinogenesis, such
as DNA adduct formation, mutagenesis, free radical scavenging, cell proliferation
and differentiation, as well as angiogenesis [14]. A matched case-control study in a
hospital was conducted to explore the association between dietary Allium consump-
tion and breast cancer risk among Iranian women and it shows that high consump-
tion of certain Allium vegetables, particularly garlic and leek, can reduce the risk of
breast cancer [15]. In rodents, garlic and its components have been reported to
inhibit the development of chemically induced tumors in the liver, thus showing
tumor cell growth inhibition and chemopreventive effects [16, 17]. It diminishes the
tumor cell growth in the prostate, bladder, and stomach [18–20].
Extracts of garlic and its related phytochemicals have anti-inflammatory poten-
tial also. One study reported that garlic extracts markedly affected liver inflamma-
tion and damage caused by Eimeria papillata infections [21], and a meta-analysis
of ten trials and one observational study found that garlic intake of 2–2.4 g/day for
four weeks or longer decreased levels of the inflammatory mediators tumor necrosis
factor-alpha (TNF-α), highly sensitive C-reactive protein (CRP), and interleukin
(IL)-6, supporting the use of garlic as an adjuvant treatment for metabolic diseases
[22]. The anti-inflammatory activity of garlic is caused by the inhibition of the emi-
gration of neutrophilic granulocytes in the epithelia. Aged black garlic (ABG) has
shown potent antioxidant activities and these activities may be responsible for its
anti-inflammatory activity [23, 24].
1498 E. Vatsa and M. Aggarwal
Aloe vera
1.2
Aloe barbadensis Miller commonly known as Aloe vera is the most popular natural
product in the prevention and ailment of various health problems and maladies. It
belongs to the family Asphodelaceae (Liliaceae). It is native to subtropical regions
and tropical climates. There are more than 400 species of Aloe belonging to family
Liliaceae. The main characteristic of the A. vera plant is its high water content,
which ranges between 99 and 99.5% [26]. The remaining 0.5–1.0% solid material
contains over 75 different potentially active compounds, including water-soluble
and fat-soluble vitamins, minerals, enzymes, simple/complex polysaccharides, phe-
nolic compounds, and organic acids [27]. Distinctive mechanisms have been pro-
posed for the wound-healing effects of Aloe vera. Glucomannan, a polysaccharide
rich in mannose, and gibberellin, a growth hormone, interact with growth factor
receptors on fibroblasts, stimulating their activity and proliferation, which in turn
significantly increases collagen synthesis after topical and oral Aloe vera [28]. A
study revealed that Aloe vera-based and chitosan-based hydrogel gels exhibited
wound-healing effects and high biocompatibility with seeded mesenchymal stem
cells used for healing grade II burns in rats [50]. In a randomized controlled clinical
trial, a polyherbal cathartic capsule of Shou Hui Tong Bian, containing Polygonum
multiflorum and Aloe vera, improved arthroscopy, replacement efficacy, recovery
time, time to postoperative exhaustion of borborygmus, and abdominal distention in
postoperative patients (N = 98) after joint replacement arthroscopy compared to
conventional treatment [29]. In a meta-analysis (23 studies, N = 4023), the authors
conclude that Aloe vera may have beneficial effects in reducing pain scores and the
severity of mucocutaneous conditions, such as psoriasis, burns, and wound healing,
compared to placebo [30]. Abbasi reported that in a double-blind RCT of 28 patients,
use of a topical skin ointment containing Aloe vera, honey, and peppermint as a
dressing for skin graft donor sites was superior to petroleum jelly in reducing wound
erythema and improved treatment satisfaction [31]. In a clinical study, to verify the
effectiveness of A. vera gel compared to silver sulfadiazine 1% cream as a burn
dressing for the treatment of superficial and partial burns, burn wound healing was
significantly earlier in patients treated with A. vera than patients treated with 1%
silver sulfadiazine [32]. Polysaccharides isolated from A. vera induce matrix metal-
lopeptidase (MMP)-3 and metallopeptidase inhibitor-2 gene expression during rat
skin wound repair, which directly helps regulate the wound-healing activity of
A. vera gel in rats [33]. Aloe vera enhanced the efficacy of topical human vascular
endothelial cell transplantation on excised full-thickness skin wounds in diabetic
mice in improving angiogenesis in part by improving glycemic control. Oral admin-
istration also promoted wound healing through inhibition of MMP-2 and MMP-9
expression [45].
Clinical studies have suggested that A. vera gel may act as a safe anti-
hyperglycemic and anti-hypercholesterolemic agent for patients with type 2 diabe-
tes without any significant effects on other normal blood lipid levels or liver or
kidney function [34]. A polyherbal formulation, including Aloe vera, was tested in
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1499
1.3 Smilax
Smilax ornata, genus of flowers inside the family Smilacaceae (Liliaceae), com-
monly called as sarsaparilla, includes approximately 300 species of woody or her-
baceous vines, variously called catbriers and greenbriers. It is mainly found in
temperate, tropical, and subtropical zones worldwide. The roots of that flora had
been used for hundreds of years in Asia and the Americas as a tonic, diuretic, and
sudorific. The rhizome, roots, stems, and leaves of sarsaparilla are utilized in tradi-
tional medicine [58]. In recent years, interest in the study of the genus Smilax has
increased. The study of the genus Smilax has drawn more attention recently.
Reviews exist about the antioxidant properties described as a 2,2-diphenyl-1-picryl-
hydrazyl (DPPH•) radical scavenger. The phenolic chemicals stilbenes, flavones,
flavanones, flavonols, smilasides, smiglasides, and helionosides are responsible for
this affiliation [66].
Smilax glabra showed protective effects against lead-induced renal oxidative
stress, inflammation, and apoptosis in weaning rats and human embryonic kidney-
293 (HEK-293) cells. Hence, it is a natural antioxidant and anti-inflammatory agent
for the treatment of lead-induced nephrotoxicity [56]. Astilbin at 5.3 mg/kg reduced
joint damage in the hind paw of complete Freund’s adjuvant (CFA)-induced arthritic
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1501
rats. Astilbin exhibited remarkable inhibitory effects on TNF-α, IL-1β, and IL-6
mRNA expressions and these effects showed inhibition of cytokine production and
inflammatory response in test mice close to anti-rheumatic drug, leflunomide [61].
The methanol extract of Smilax 400 mg/kg claimed to produce anti-inflammatory
activity in the bradykinin-prompted and prostaglandin-induced edema models [65].
Kwon et al. suggested a novel molecular mechanism for water extract of Gleditsia
sinensis thorns (WESGR)-mediated anti-prostate cancer effects at particular steps
such as with migration and adhesion to collagen, and it could provide the possibility
of therapeutic utilization of WESGR against prostate cancer progression [57]. She
et al. observed that the supernatant of water-soluble extract from Smilax glabra
Roxb. (SGR) should enhance adhesion, inhibit migration and invasion of HepG2,
M.D. Anderson - Metastatic Breast 231 (MDA-MB-231), and T24 cells in vitro, as
well as diminish metastasis of MDA-MB-231 cells in vivo. Outcomes of F-actin
and vinculin dual staining showed the improved focal adhesion in SW-dealt-with
cells. Microarray evaluation indicated a repression of transforming growth factor-
beta (TGF-β1) signaling by means of SW remedy, which became verified by real-
time reverse transcription-polymerase chain reaction (RT-PCR) of TGF-β1-associated
genes and immunoblotting of transforming growth factor beta receptor I (TGFBR1)
protein [60]. Song et al. observed the growth inhibitory action of Smilax by treat-
ment of its water-soluble extract 3.5 μg/μL on multiple cancer cells in vitro and in
vivo, and redox-dependent persistent activation of extracellular signal-regulated
kinase 1 (ERK1/2) [62]. Another study revealed that SGR inhibited growth of
human breast cancer mobile line Michigan Cancer Foundation-7 (MCF7), colon
carcinoma mobile line human colorectal adenocarcinoma cell line (HT-29), and
gastric cancer cellular line human gastric cancer cell line (BGC-823) in a dose-
structured way [63].
Huang et al. obtained total flavonoids (which include four marker compounds) of
S. glabra, and the full content became as much as 55.6%. The consequences recom-
mended that total flavonoids of S. glabra (TFSG) has huge effect on reducing uric
acid in hyperuricemic mice by means of inhibiting the xanthine oxidase (XOD)
sports and upregulating the expression of renal organic anion transporter 1 (OAT1)
and organic cation transporter novel family member 2 (OCTN2) and their mRNA in
kidney tissue of hyperuricemic mice [59].
A study revealed that SGR extract inhibited the HepG2 and Hep3B cell growth
by causing cell-cycle arrest at either S phase or S/G2 transition and induced apop-
tosis, evidenced by a DNA fragmentation assay [64]. In vitro assay demonstrated
the antioxidant potential of Smilax. The Smilax triggered a big reduction of the
formalin-evoked flinches in rats, an effect reversed through opioid antagonist nalox-
one [66]. Rafatullah et al. studied the effect of ethanol extract of sarsaparilla 100 mg/
kg/day for 90 days on carbon tetrachloride (CCl4)-induced hepatocellular damage
in rats [67].
Nithyamala et al. investigated analgesic interest of root powder of Smilax by
means of hot plate technique and acetic acid prompted writhing technique in albino
mice. The oral administration of root substantially expanded the response time in a
dose-based manner in warm plate technique. The basis powder also induced
1502 E. Vatsa and M. Aggarwal
inhibitory impact on writhing triggered by using acetic acid [68]. Methanolic extract
of Smilax roots examined for its immunomodulatory interest by means of nitro blue
tetrazolium chloride (NBT) reduction test and anti-arthritic test by in vitro protein
denaturation and in vivo complete Freund’s adjuvant (CFA) precipitated arthritis.
Extract at 200 mg/kg and 400 mg/kg showed statistically significant inhibition
(p < 0.05) of the edema formation in CFA model [69].
Leaf and fruit extracts of Smilax were shown to exhibit antimicrobial and anti-
oxidant activities, which may be attributed to the presence of secondary metabolites
such as alkaloids, flavonoids, tannins, triterpenoids, and sterols [73]. In vitro anti-
oxidant activities of leaf and stem extracts of Smilax were performed, which revealed
the reducing power of leaf and stem extracts of Smilax [70]. Another study revealed
marked reducing activity of methanolic extract of stem when compared to aqueous
extract of stem [71]. Muthu et al. investigated the evaluation of in vivo antioxidant
activity of ethanolic extract of root on aluminum-chloride-induced apoptosis sup-
pressing antioxidative stress in Wistar rats and found that Smilax can be used as an
antioxidant that is beneficial in preventing the progress of various oxidative
stresses [72].
1.4 Meadow Saffron
acute gout in a 51-year-old patient with history of recurrent acute episodes of arthri-
tis [82]. In a randomized placebo-controlled trial evaluating the efficacy and toxicity
of colchicine in acute gout, patients were treated with an initial dose of 1 mg fol-
lowed by sequential doses of 0.5 mg every two hours until either significant symp-
tomatic relief or intolerance. Patients treated with colchicine were observed to
experience pain relief within 48 hours as compared to placebo [86]. The AGREE—
Acute Gout Flare Receiving Colchicine Evaluation—trial was the first randomized,
placebo-controlled trial to compare low- and high-dose colchicine and it was
observed that there is a comparable response rate in the low-dose group versus high-
dose group [76]. Also, high-dose group showed more side effects like diarrhea and
nausea [76]. A placebo-controlled randomized trial evaluating colchicine for flare
prophylaxis in patients with chronic gout initiating allopurinol found that subjects
treated with colchicine had fewer and less severe flares [87].
In a randomized controlled clinical trial, two groups were divided: the patients in
the intervention and control groups were treated with Rhazes tablet + Ibuprofen
pearl (400 mg) PRN (pro re nata) and placebo tablet + Ibuprofen pearl (400 mg)
PRN, respectively. It was concluded that Rhazes tablet can be used as a pharmaco-
logical intervention to reduce pain in patients with lower back pain [83].
Since 1974, colchicine is the treatment of choice for Familial Mediterranean
Fever (FMF). It is evident through a randomized controlled trial in which patients
were treated with colchicine and fewer attacks were observed [88]. A randomized
cohort study showed favorable efficacy of colchicine in treatment of secondary
amyloidosis [89]. Colchicine was found more effective than melphalan and predni-
sone in increasing the survival rate of patients [89].
Colchicine was reported to be used in Behçet’s syndrome; 7–12 patients were
reported to show improvement in symptoms when treated with colchicine 0.5 mg
twice a day [90]. A double-blind controlled trial wherein 116 sufferers with Behçet’s
syndrome were randomized to get hold of either colchicine or placebo for 2 years
showed enormous reductions in the occurrence of genital ulcers, erythema nodo-
sum, and arthritis in woman patients, in addition to a decrease in the incidence of
arthritic signs in men [91, 92].
All parts of the plant, especially the bulbs, are highly toxic. If taken in abundance
it can be highly toxic and can be fatal. Cramping, vomiting, diarrhea, increased
blood pressure, and respiratory failure are the poison symptoms [93]. Formerly,
poisoning occurred when people used homemade preparations. Due to this, many
deaths have been reported [94].
1.5 Asparagus
bioactive constituents of asparagus are a group of steroidal saponins. This plant also
contains vitamins A, B1, B2, C, and E, as well as Mg, P, Ca, Fe, and folic acid. Other
primary chemical constituents of asparagus are essential oils, asparagine, arginine,
tyrosine, flavonoids (kaempferol, quercetin, and rutin), resin, and tannin [97, 98].
Saponins have a wide range of biological activities, including those of antioxidants,
immunostimulants, antihepatotoxic, antibacterial, beneficial for diabetic retinopa-
thy, anticarcinogenic, antimicrobials, that inhibit molds, antidiarrheal, and antiul-
cerogenic agents [97, 99].
The hypoglycemic effects of asparagus extracts were evaluated by streptozoto-
cin (STZ)-induced diabetic rats, which had the same efficacy at a dose of 500 mg/
kg as the antidiabetic drug glibenclamide (5 mg/kg rat body weight) [100].
Asparagus juice (CAJ) from asparagus old stem was used in type I diabetic rat
model, and results showed that CAJ reduced the blood glucose level along with
lipid level in diabetic rats by decreasing the content of serum glucose, total choles-
terol, and MDA, and improved level of serum insulin [101]. Hypoglycemic activity
of asparagus old stem was also reported by Zhao (2010). He reported the presence
of flavonoids, polyphenols, saponins, and polysaccharides, which showed remark-
able hypoglycemic effects at 1.43, 5.58, 1.82, and 4.24 mg/g dry weight, respec-
tively [102]. The extract of A. officinalis in the diabetic rats showed potent
antioxidant activity and improvement in β-cell function both at 250 mg/kg and
500 mg/kg. The insulin–glucose ratio was reported to increase at both doses [103].
A. officinalis also shows antitumorigenic and antimetastatic effects. In a trans-
genic mouse model of high-grade serous ovarian cancer, asparagus was reported to
decrease the cellular viability and caused cell-cycle G1 phase arrest. It inhibited
tumor growth and increased phosphorylation of adenosine monophosphate-acti-
vated protein kinase (AMPK) in the ovarian tumor tissues [104]. The inedible bot-
tom part of asparagus was utilized as a supplement and the saponins from old stem
of asparagus suppressed the cell viability of breast, colon, and pancreatic cancers.
The extract inhibited the tumor cell motility [105]. The popular vegetable dish of
asparagus, i.e., the shoots of white asparagus, was reported to possess antioxidant,
anti-inflammatory, and antitumor activities. The Wistar rats with induced colon car-
cinogenesis were treated with asparagus for seven weeks and the rats exhibited a
50% reduction in the amount of pre-neoplastic lesions and promoted normal cellu-
lar homeostasis [106]. Xiang et al. also reported the anticancer effects of asparagus.
Deproteinized asparagus polysaccharide exhibited widespread anticancer activity in
opposition to hepatocellular carcinoma cells and sensitized the tumoricidal conse-
quences of mitomycin, indicating that asparagus is a chemosensitizer for liver can-
cer therapy [107]. Mechanistic studies revealed the inhibition of migration, invasion,
and angiogenesis of cancer cells [108].
Asparagus showed antioxidant, anti-inflammatory, and antihepatotoxic proper-
ties in the 40 Wistar rats that were given 400 mg/kg of the extract. Asparagus extract
increased the total antioxidative capability and improved function of liver and kid-
ney tissues. According to the researchers, it has a potential protective action against
oxidative stress, and liver and kidney damage [109]. Also, asparagus seed extract
additionally accelerated general antioxidant reputation at a dose of 500 mg/kg in
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1505
streptozotocin-induced diabetic rats [100]. Ku et al. (2017) reported the high bind-
ing capacity of soluble polyphenols of asparagus to human serum albumin and it
also showed the protective effects against bisphenol-A-induced toxicity [110].
Asparagus has potential against bile acids, which are the important excretory
pathways for cholesterol metabolism. Asparagus effect was studied on cholesterol
as it reduced the simulating gastrointestinal digestion and measured binding capac-
ity of the food and bile acid [98]. Another study indicated the cholesterol-lowering
activity of asparagus in hyperlipidemia mouse model; polysaccharides present on
asparagus lowered the lipoprotein cholesterol [111, 112]. Asparagus also showed
the positive results in lowering the blood pressure from human clinical trials and
therefore can be used as an antihypertensive agent [113].
There are various other therapeutic as well as physiological effects of asparagus.
The instant powder of asparagus increased the sleep time in insomnia patients, and
also shortened the sleep time through the food test that was performed on 60 volun-
teers [114]. Jager et al. (2005) proved the antiepileptic activity of asparagus instant
powder [115] and Miura et al. (2016) discovered the stress-relieving property [116].
Lily of the valley is a common name of Convallaria majalis. This plant belongs to
Liliaceae family. The plant contains numerous steroidal glycosides, cardiac glyco-
sides, flavonoids, chelidonic acid, choline, and azetidine-2-carbonic acid. In rhi-
zomes and roots of the plant, steroidal saponins are predominant [117–120]. From
ancient times, lily of the valley is used for the treatment of various cardiovascular
ailments including congestive heart failure, cardiomyopathy, irregular heartbeat,
urinary tract infections (UTIs), kidney stones, epilepsy, edema, and various eye
infections. It is also used in the treatment of leprosy [121]. An ointment made from
the roots is used in the treatment of burns and to prevent scar tissue. However, owing
of the plant’s potential toxicity, it should never be used without first consulting a
professional [131].
Cardiac glycosides, also known as cardenolides, are a type of steroid that has
long been known to have a positive ionotropic impact on the heart. All cardiac gly-
cosides increase intracellular sodium by affecting ion transport across cardiac mus-
cle cell membranes via effects on Na(+)/K(+)-ATPase enzymes. As a result,
intracellular calcium levels rise, improving heart contractility. Atrial flutter and
fibrillation are also converted to steady sinus rhythm with cardiac glycosides
[123, 124].
Convallaria majalis has been demonstrated in animal trials to raise K+ in the
atria and atrial stroke volume. Despite the fact that convallatoxin is a vasoconstric-
tor, the sum of all cardiac glycosides and other ingredients may have a greater vaso-
tonic effect, improving circulation and coronary flow [125, 126].
A study was done for the determination of whether convallatoxin present in the
plant as a cardiac glycoside induces the expression of tissue factor and leads to
1506 E. Vatsa and M. Aggarwal
hypercoagulable state. The findings indicate that the convallatoxin induces the tis-
sue factor expression in endothelial cells and also induces hypercoagulable
state [122].
Convallaria majalis is a homoeopathic medicine that has a beneficial effect on
the heart and also acts as a diuretic. Lateef et al. (2010) evaluated the effect of lily
of the valley on kidney function by investigating the effect of plant extract on serum
uric acid and creatinine on rabbits for 14 days. After the completion, animals were
sacrificed to collect blood sample. It was concluded that the alcoholic extract at
10 mg/kg acts as a significant hypouricemic agent [127].
Another study indicated the effect of convallamaroside, i.e., the steroidal saponin
present in Convallaria majalis. Isolated extract was injected to the mice and evalu-
ated by intradermal test. Convallamaroside medication to mice reduced the number
of new vasculatures produced by sarcoma mouse cells (p = 0.001) [128].
For a single ingredient, convallatoxol, the dose that killed 50% of a sample popu-
lation (LD50) in cats was 0.14 mg/kg given intraperitoneally [130]. This herb’s
human dose is 0.6 g powdered herb or 5–10 drops tincture, split daily. A case report
detailed a dog that died abruptly after consuming lily of the valley in its enclosed
yard [129].
The essential oil of lily of the valley is used in aromatherapy to treat headaches,
depression, and melancholy. Memory loss, apoplexy, and epilepsy can all be treated
with it. It is utilized to help brain cells grow stronger and to boost cognitive func-
tions. UTI is treated with a tincture made from lily of the valley flowers, which
clears blockages from the urethra. Because of its purgative and laxative properties,
this herb is commonly used as a substitute for aloes. This, in turn, keeps the diges-
tive system running smoothly.
The lily of the valley also has the following advantages:
• Kidney stones are broken down.
• Prevents the body from retaining water.
• Pain associated with joint tissues such as gout and rheumatism are reduced.
• Conjunctivitis is treated with this medication.
• Paralysis, shock, and speech loss are all treated using essential oils.
• Aids in the treatment of leprosy and edema.
• Poisoning and drunkenness are treated by producing vomiting.
2 Conclusions
Natural products act as very potential products for new drug discovery and have the
ability to generate new drug leads that will be further subjected for clinical trials. In
the present study, several advances in the field of technology of natural products
were discussed and how they are subjected for clinical trials were described in
detail. In this chapter, several conclusive points were also discussed on the basis of
analyzed data and therapeutic-evidence-based studies, which provide a very strong
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1507
References
lial function in diabetes: a randomized, double-blind, placebo-controlled trial. Exp Ther Med
19(2):1485–1489. https://doi.org/10.3892/etm.2019.8377
12. Liu J, Zhang G, Cong X, Wen C (2018) Black garlic improves heart function in patients with
coronary heart disease by improving circulating antioxidant levels. Front Physiol 9:1435.
https://doi.org/10.3389/fphys.2018.01435
13. Gebhardt R, Beck H (1996) Differential inhibitory effects of garlic-derived organosul-
fur compounds on cholesterol biosynthesis in primary rat hepatocyte cultures. Lipids
31(12):1269–1276. https://doi.org/10.1007/bf02587912
14. Capasso A (2013) Antioxidant action and therapeutic efficacy of Allium sativum L. Molecules
18(1):690–700
15. Pourzand A, Tajaddini A, Pirouzpanah S, Jafarabadi MA, Samadi N, Ostadrahimi AR, Sanaat
Z (2016) Associations between dietary Allium vegetables and risk of breast cancer: a hospital-
based matched case-control study. J Breast Cancer 19(3):292–300. https://doi.org/10.4048/
jbc.2016.19.3.292
16. Kweon S, Park KA, Choi H (2003) Chemopreventive effect of garlic powder diet in diethyl-
nitrosamine induced rat hepatocarcinogenesis. Life Sci 73(19):2515–2526
17. Knowles LM, Milner JA (2003) Diallyl disulfide induces ERK phosphorylation and alsters
gene expression profiles in human colon tumor cells. J Nutr 133(9):2901–2906. https://doi.
org/10.1093/jn/133.9.2901
18. Hsing AW, Chokkalingam AP, Gao YT, Madigan MP, Deng J, Gridley G, Fraumeni JF (2002)
Allium vegetables and risk of prostate cancer: a population-based study. J Natl Cancer Inst
94(21):1648–1651. https://doi.org/10.1093/jnci/94.21.1648
19. Lau BH, Woolley JL, Marsh CL, Barker GR, Koobs DH, Torrey RR (1986) Superiority of
intralesional immunotherapy with Corynebacterium parvum and Allium sativum in control of
murine transitional cell carcinoma. J Urol 136(3):701–705
20. Wattenberg LW, Sparnis VL, Barany G (1989) Inhibition of N-nitrosodiethylamine carcino-
genesis in mice by naturally occurring organosulfur compounds and monoterpenes. Cancer
Res 49(10):2689–2692
21. Ahmad TA, Sayed BA, Sayed LH (2016) Development of immunization trials against Eimeria
spp. Trials Vaccinol 5:38–47. https://doi.org/10.1016/j.trivac.2016.02.001
22. Koushki M, Dashatan NA, Pourfarjam Y, Doustimotlagh AH (2021) Effect of garlic intake on
inflammatory mediaors: a systematic review and meta-analysis of randomized controlled tri-
als. Postgrad Med J 97(1145):156–163. https://doi.org/10.1136/postgradmedj-2019-137267
23. Hobauer R, Frass M, Gmeiner B, Kaye AD, Frost EA (2000) Garli extract (allium sati-
vum) reduces migration of neutrophils through endothelial cell monolayers. Middle East J
Anaesthesiol 15(6):649–658
24. Gu X, Wu H, Fu P (2013) Allicin attenuates inflammation and suppresses HLA-B27 pro-
tein expression in ankylosing spondylitis mice. Biomed Res Int:171573. https://doi.
org/10.1155/2013/171573
25. Chan JY, Yuen AC, Chan RY, Chan SW (2013) A review of the cardiovascular benefits and
antioxidant properties of allicin. Phytoher Res 27(5):637–646
26. Hamman JH (2008) Composition and application of Aloe vera leaf gel. Molecules
13:1599–1616. https://doi.org/10.3390/molecules13081599
27. Maharjan HR, Nampoothiri PL (2015) Evaluation of biological properties and clinical effec-
tiveness of Aloe vera: a systematic review. J Tradit Complement Med 5(1):21–26. https://doi.
org/10.1016/j.jtcme.2014.10.006
28. Chithra P, Sjithlal GB, Chandrakasen G (1998) Influence of Aloe vera on collagen character-
istics in healing dermal wounds in rats. Mol Cell Biochem 181(1–2):71–76. https://doi.org/1
0.1023/a:1006813510959
29. Huang S, Xie Y, Huang Z, Zhang G, Chen G, Yuan J, Wang J, Xiangyang L, Zhou Y (2021)
Effect of observation of Shou Hui Tong Bian Capsule (Polygonum Multiflorum and Aloe-
Based Herbal Capsule for Cathartic Effect) in rapid rehabilitation of joint surgery. Evi Based
Compl Alternat Med:2268464. https://doi.org/10.1155/2021/2268464
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1509
30. Metin ZG, Helvaci A, Eren MG (2021) Effects of Aloe vera in adults with mucocutaneous
problems: a systematic review and meta-analysis. J Adv Nurs 77(3):1105–1126. https://doi.
org/10.1111/jan.14653
31. Abbasi MS, Rahmati J, Ehsani AH, Takzare A, Partoazar A, Takzaree N (2020) Efficacy of
a natural topical skin ointment for managing Split-thickness skin graft donor sites: a pilot
double-blind randomized controlled trial. Adv Skin Wound Care 33(7):1–5. https://doi.
org/10.1097/01.asw.0000666916.00983.64
32. Shahzad MN, Ahmed N (2013) Effectiveness of Aloe vera gel compared with 1% silver
sulphadiazine cream as burn wound dressing in second degree burns. J Pak Med Assoc
63:225–230
33. Tabandeh MR, Oryan A, Mohammadalipour A (2014) Polysaccharides of Aloe vera induve
MMP-3 and TIMP-2 gene expression during the skin wound repair of rat. Int J Biol Macromol
65:424–430
34. Huseini HF, Kianbakht S, Hajiaghaee R, Dabaghian FH (2012) Anti-hyperglycemiac and
anti-hypercholesterolemic effects of Aloe vera leaf gel in hyperlipidemic type 2 diabetic
patients: a randomized double-blind placebo-controlled clinical trial. Planta Med 78:311–316
35. Cholesterol Treatment Trialists’ (CTT) Collaborators, Kearney PM, Blackwell L, Collins R,
Keech A, Simes J, Peto R, Armitage J, Baigent C (2008) Efficacy of cholesterol-lowering
therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis.
Lancet. 371:117–125. https://doi.org/10.1016/S0140-6736(08)60104-X
36. Devaraj S, Jialal R, Jialal I, Rockwood R (2008) A pilot randomized placebo-controlled trial
of 2 Aloe vera supplements in patients with pre-diabetes/metabolic syndrome. Planta Med
p. 74–77
37. Jain N, Vijayaraghavan R, Pant SC, Lomash V, Ali m. (2010) Aloe vera gel alleviates cardio-
toxicity in streptozocin-induced diabetes in rats. J Pharm Pharmacol 62:115–123
38. Rahimifard M, Nigjeh MN, Mahroui N (2013) Improvement in the function of isolated rat
pancreatic islets through reduction of oxidative stress using traditional Iranian medicine. Cell
J:147–163
39. Hamiza OO, Rehman MU, Khan R (2014) Chemopreventive effects of aloin against
1,2-dimethylhydrazine-induced preneoplastic lesions in the colon of Wistar rats. Hum Exp
Toxicol 33:148–163. https://doi.org/10.1177/0960327113493307
40. Kim HS, Lee BM (1997) Inhibiting of benzo[a]pyrene-DNA adduct formation by Aloe bar-
badensis miller. Carcinogenesis 18(4):771–776
41. Kim hs, Kacew S, Lee BM. (1999) In vitro chemopreventive effects of plant polysaccharides
(Aloe barbadensis miller, Lentinus edodes, Ganoderma lucidum and Coriolus versicolor).
Carcinogenesis 20(8):1637–1640
42. Babaee N, Zabihi E, Mohseni S, Moghadamnia AA (2012) Evaluation of the therapeutic
effects of Aloe vera gel on minor recurrent aphthous stomatitis. Dent Res J 9:381–385
43. Mansour G, Ouda S, Shaker A, Abdallah HM (2014) Clinical efficacy of new Aloe vera-
myrrh-based oral mucoadhesive gels in the management of minor recurrent aphthous stoma-
titis: a randomized, double-blind, vehicle-controlled study. J Oral Pathol Med 43:405–409
44. Najafian Y, Khorasani ZM, Njafi MN, Hamedi SS, Mhjour M, Feyzabadi Z (2019)
Efficacy of Aloe vera/Plantago major gel in diabetic foot ulcer: a randomized double-
blind clinical trial. Curr Drug Discov Technol 16(2):223–231. https://doi.org/10.217
4/1570163815666180115093007
45. Kaewsrisung S, Sukpat S, Issarasena N, Patumraj S, Somboonwong J (2021) The effects
of oral Aloe vera on the efficacy of transported human endothelial cells and the expression
of matrix metalloproteinases in diabetic wound healing. Heliyon 7(12):e08533. https://doi.
org/10.1016/j.heliyon.2021.e08533
46. Farid A, Tawfik A, Elsioufy B, Safwat G (2021) In vitro and in vivo anti-cryptosporidium and
anti-inflammatory effects of Aloe vera gel in dexamethasone immunosuppressed mice. Int J
Parasitol Drugs Drug Resis:156–167. https://doi.org/10.1016/j.ijpddr.2021.09.002
1510 E. Vatsa and M. Aggarwal
47. Liu C, Hua H, Zhu H, Cheng Y, Guo Y, Yao W, Qian H (2021) Aloe polysaccharides amelio-
rate acute colitis in mice via Nrf2/HO-1 signaling pathway and short-chain fatty acids metab-
olism. Int J Biol Macromol 185:804–812. https://doi.org/10.1016/j.ijbiomac.2021.07.007
48. An J, Lee H, Lee S, Song Y, Kim J, Park IH, Kong H, Kim K (2021) Modulation of pro-
inflammatory and anti-inflammatory cytokines in the fat by an aloe gel-based formula,
QDMC, is correlated with altered gut microbiota. Immune Netw 21(2):e15. https://doi.
org/10.4110/in.2021.21.e15
49. Akgul KT, Dogantekin E, Ozer E, Kotanoglu M, Gokkurt Y, Hucumenoglu S (2021)
Histopathological effects of aloe vera on wound healing: an experimental study. Turk J Med
Sci 51(4):2193–2197. https://doi.org/10.3906/sag-2102-224
50. Sharifi E, Chehelgerdi M, Kelishadrokhi AF, Nafchi FY, Dehkordi KA (2021) Composition
of therapeutic effects of encapsulated Mesenchymal stem cells in Aloe vera gel and Chitosan-
based gel in healing of grade-II burn injuries. Regen Ther 18:30–37. https://doi.org/10.1016/j.
reth.2021.02.007
51. Koga AY, Felix JC, Silvestre RGM, Lipinski LC, Carletto B, Kawahara FA, Pereira AV
(2020) Evaluation of wound healing effect of alginate film containing Aloe vera gel and
cross-linked with zinc chloride. Acta Cir Bras 35(5):e202000507. https://doi.org/10.1590/
s0102-865020200050000007
52. Sumi FA, Sikder B, Rahman MM, Lubna SR, Ula A, Hossain MH, Jahan IA, Alam MA,
Subhan N (2019) Phenolic content analysis of Aloe vera gel and evaluation of the effect of
aloe gel supplementation on oxidative stress and fibrosis in isoprenaline-administered cardiac
damage in rats. Prev Nutr Food Sci 24(3):254–264. https://doi.org/10.3746/pnf.2019.24.3.254
53. Gupta VK, Siddiqi NJ, Ojha AK, Sharma B (2019) Hepatoprotective effect of Aloe
vera against cartap- and malathion-induced toxicity in Wistar rats. J Cell Physiol
234(10):18329–18343. https://doi.org/10.1002/jcp.28466
54. Sehitoglu MH, Karaboga I, Kiraz A, Kiraz HA (2018) The hepatoprotective effect of Aloe
vera on ischemia-reperfusion injury in rats. North Clin Istanb 6(3):203–209. https://doi.
org/10.14744/nci.2018.82957
55. Jung E, Kim J (2018) Aloin inhibits muller cells swelling in a rat model of thioacetamide-
induced hepatic retinopathy. Molecules 23(11):2806. https://doi.org/10.3390/
molecules23112806
56. Shi Y, Tian C, Yu X, Fang Y, Zhao X, Xia D (2020) Protective effects of Smilax glabra Roxb.
Against Lead-induced renal oxidative stress, inflammation and apoptosis in weaning rats and
HEK-293 cells. Front Pharmacol. https://doi.org/10.3389/fphar.2020.556248
57. Kwon OY, Ryu S, Choi JK, Lee SH (2020) Smilax glabra Roxb. Inhibits collagen induced
adhesion and migration of PC3 and LNCaP prostate cancer cells through the inhibition of Beta
1 integrin expression. Molecules 25(13):3006. https://doi.org/10.3390/molecules25133006
58. Saleem SA, Lakshmi B, Kumar JNS (2021) Review article on traditional out look of Smilax
zeylanica. Res J Pharmacol Pharmacodyn. https://doi.org/10.52711/2321-5836.2021.00014
59. Huang L, Deng J, Chen G, Zhou M, Liang J, Yan B, Shu J, Liang Y, Huang H (2019) The
anti-hyperuricemic effect of four astilbin stereoisomers in S.glabra on hyperuricemic mice. J
Ethnopharmacol. https://doi.org/10.1016/j.jep.2019.03.004
60. She T, Zhao C, Feng J, Wang L, Qu L, Fang K, Cai S, Shou C (2015) Sarsaparilla (Smilax
glabra Rhizome) extract inhibits migration and invasion of cancer cells by suppressing TGF-
β1 pathway. PLoS One 10(3):e0118287. https://doi.org/10.1371/journal.pone.0118287
61. Dong L, Zhu J, Du H, Nong H, He X, Chen X (2017) Astilbin from Smilax glabra Roxb.
Attenuates inflammatory responses in complete freund’s adjuvant-induced arthritis rats. Evid
Based Complement Alternat Med:8246420. https://doi.org/10.1155/2017/8246420
62. Song G, She T, Feng J, Lian S, Li R, Zhao C, Luo J, Dawuti R, Cai S, Qu L, Shou C (2017)
Sarsaparilla (Smilax glabra rhizome) extract activates redox-dependent ATM/ATR path-
way to inhibit cancer cell growth by Sphase arrest, apoptosis and autophagy. Nutr Cancer
69(8):1281–1289. https://doi.org/10.1080/01635581.2017.1362447
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1511
63. Gao Y, Su Y, Qu L, Xu S, Meng L, Cai SQ, Shou C (2011) Mitochondrial apoptosis contrib-
utes to the anti-cancer effect of Smilax glabra Roxb. Toxicol Lett 207(2):112–120. https://doi.
org/10.1016/j.toxlet.2011.08.024
64. Sa F, Gao JL, Fung KP, Zheng Y, Lee SMY, Wang YT (2008) Anti-proliferative and pro-
apoptotic effect of Smilax glabra Roxb. Extract on hepatoma cell lines. Chem Biol Interact
171(1):1–14. https://doi.org/10.1016/j.cbi.2007.08.012
65. Khan A, Singh PD, Reese PB, Howden J, Golding M, Thomas TT (2020) Investigation of
the preliminary mechanism of action for the acute anti-inflammatory activity of the metha-
nol extract of Smilax ornate Lem. J Ethanopharmacol 248:112360. https://doi.org/10.1016/j.
jep.2019.112360
66. Hirota BCK, Paula CDS, Oliveira VBD, Cunha JMD, Schreiber AK, Ocampos FMM, Barison
A, Miguel OG, Miguel MD (2016) Phyochemical and antinociceptive, anti-inflammatory and
anti-oxidant studies of Smilax larvata(Smilacaceae). Evi Based Comp Alt Med. https://doi.
org/10.1155/2016/9894610
67. Rafatullah S, Mossa JS, Ageel AM, Yahya MAA (2008) Hepatoprotective and
safety evaluation studies on sarsaparilla. Pharm Biol 29(4):296–301. https://doi.
org/10.3109/13880209109082901
68. Nithyamala I, Ayyasamy S, Pitchiahkumar M, Kumar A, Velapandian V (2013) Evaluation
of analgesic and antiinflammatory activity of siddha drug Karuvilanchiverchoornam (root
powder of Smilax zeylanica Linn) in rodents. IOSR J Pharm Biol Sci 6(1):6–11
69. Babu PV, Ashwini T, Krishna MV, Raju MG (2017) Immunomodulatory and antiarthritic
activities of smilax zeylanica. Int J Phytomed 9(1). https://doi.org/10.5138/09750185.1963
70. Uddin MN, Ahmed T, Pathan S, Al-Amin MM, Rana MS (2015) Antioxidant and cytotoxic
activity of stems of Smilax zeylanica in vitro. J Basic Clin Physiol Pharmacol 26(5):453–463
71. Thirugnanasampandan R, Mutharaiah VN, Bai NV (2009) In vitro propagation and free radi-
cal studies of Smilax zeylanica vent. Afr J Biotechnol 8(3):395–400
72. Muthu AK, Senthil BS, Kalaichelvan VK (2018) In vivo antioxidant activity of ethanolic
extract from root of smilax on aluminium chloride induced oxidative stress in wistar rats. J
Drug Delivery Therapeutics 8(6). https://doi.org/10.22270/jddt.v8i6-s.2078
73. Shree VSD, Arbin A, Noorain GKS, Sahana BK, Kekuda TRP (2018) Preliminary phy-
tochemical analysis, antimicrobial and antioxidant activity of Smilax. J Drug Delivery
Therapeutics. 8(4). https://doi.org/10.22270/jddt.v8i4.1779
74. Toplan GG, Gurer C, Mat A (2016) Importance of colchicum species in modern therapy and
its significance in Turkey. J Fac Pharm 46:129–144
75. Alali FQ, Tawaha K, El-Elimat T (2007) Determination of (−)-demecolcine and (−)-colchi-
cine content in selected Jordanian Colchicum species. Pharmazie 62(10):739–742
76. Terkeltaub RA, Furst DE, Bennett K, Kook KA, Crockett RS, Davis MW (2010) High versus
low dosing of oral colchicine for early acute gout flare: twenty-four-hour outcome of the first
multicenter, randomized, double-blind, placebo-controlled, parallel-group, dose-comparison
colchicine study. Arthritis Rheum 62(4):1060–1068. https://doi.org/10.1002/art.27327
77. Sakane T, Takeno M (2000) Novel approaches to Behcet’s disease. Expert Opin Investig
Drugs 9(9):1993–2005
78. Cifuentes M, Schilling B, Ravindra R, Winter J, Janik ME (2006) Synthesis and biologi-
cal evaluation of B-ring modified colchicine isocolchicineanalogs. Bioorg Med Chem Lett
16(10):2761–2764. https://doi.org/10.1016/j.bmcl.2006.02.010
79. Cocco G, Chu DC, Pandolfi S (2010) Colchicine in clinical medicine. A guide for internists.
Eur J Intern Med 21(6):503–508. https://doi.org/10.1016/j.ejim.2010.09.010
80. Ondra P, Valka I, Sutlupinar N, Simanek V (1995) Chromatographic determination of con-
stituents of the genus colchicum (Liliaceae). J Chromatogr A 704:351–356
81. Akram M, Alam O, Khan U, Naveed A, Asif HM (2012) Colchicum autumnale:a review.
Acad J 6:8. https://doi.org/10.5897/JMPR.9000370
82. Mikkelsen WM, Salin RW, Duff IF (1956) Alopecia totalis after desacetylmethylcolchicine
therapy of acute gout-report of a case. N Engl J Med 255:769–770
1512 E. Vatsa and M. Aggarwal
83. Estebanez EB, Alconero LL, Fernandez BJ, Marguello MG, Caro JCL, Vallejo JD, Sampedro
MF, Cacho PM, Espiga CR, Saiz MMG (2021) The effectiveness of early colchicine
administration in patients over 60 years old with high risk of developing severe pulmonary
complications associated with coronavirus pneumonia SARS-CoV-2 (COVID-19): study
protocol for an investigator-driven randomized controlled clinical trial in primary health
care—COLCHICOVID study. Trials 22:590–592
84. Malek VG, Parvari S, Jafari F, Rouhani Y, Rahimi R, Abbassian A (2019) Efficacy of a tra-
ditional herbal formula based on Colchicum autumnale L. (Rhazes tablet) in low back pain:
a randomized controlled clinical trial. Int J Ayurvedic Med 10(1). https://doi.org/10.47552/
ijam.v10i1.1178
85. Slobodnick A, Shah B, Pillinger MH, Krasnokutsky S (2015) Colchicine: old and new. Am J
Med 128(5):461–470. https://doi.org/10.1016/j.amjmed.2014.12.010
86. Ahem MJ, Reid C, Gordon TP, McCredie M, Brooks PM, Jones M (1987) Does colchicine
work? The results of the first controlled study in acute gout. Aust NZ J Med 17(3):301–304.
https://doi.org/10.1111/j.1445-5994.1987.tb01232.x
87. Borstad GC, Bryant LR, Abel MP, Scroggie DA, Harris MD, Alloway JA (2004) Colchicine
for prophylaxis of acute flares when initiating allopurinol for chronic gouty arthritis. J
Rheumatol 31(12):2429–2432
88. Dinarello CA, Wolff SM, Goldfinger SE, Dale DC, Alling DW (1974) Colchicine therapy for
familial Mediterranean fever. A double-blind trial. N Engl J Med 291(18):934–937. https://
doi.org/10.1056/nejm197410312911804
89. Kyle RA, Gertz MA, Greipp PR, Witzig TE, Lust JA, Lacy MQ, Therneau TM (1997) A
trial of three regimens for primary amyloidosis: colchicine alone, melphalan and prednisone
and melphalan, prednisone and colchicine. N Engl J Med 336(17):1202–1207. https://doi.
org/10.1056/nejm199704243361702
90. Matsumura N, Mizushima Y (1975) Leucocyte movement and colchicine treatment in
Behcet’s disease. Lancet 2(7939):813. https://doi.org/10.1016/s0140-6736(75)80031-6
91. Komlodi-Pasztor E, Sackett DL, Fojo AT (2012) Inhibitors targeting mitosis: tales of how
great drugs against a promosing target were brought down by a flawed rationale. Clin Cancer
Res 18(1):51–63. https://doi.org/10.1158/1078-0432.ccr-11-0999
92. Yurdakul S, Mat C, Tuzun Y, Ozyazgen Y, Hamuryudan V, Uysal O, Senocak M, Yazici H (2001)
A double-blind trial of colchicine in Behcet’s syndrome. Arthritis Rheum 44(11):2686–2692.
https://doi.org/10.1002/1529-0131(200111)44:11<2686::aid-art448>3.0.co;2-h
93. Poutaraud A, Girardin P (2002) Alkaloids in Meadow Saffron, Colchicum autumnalel. J
Herbs Spices Med Plants 9(2):63–79. https://doi.org/10.1300/J044v09n01_08
94. Dasgeb B, Kornreich D, McGuinn K, Okon L, Brownell I, Sackett DL (2019) Colchicine: an
ancient drug with novel applications. Br J Dermatol 178(2):350–356. https://doi.org/10.1111/
bjd.15896
95. Gaur RD (1999) The flora of the district Garhwal North West Himalaya. TransMedia,
Srinagar, Garhwal, p 170
96. Kirtikar KR, Basu BD (1984) Indian medicinal plants. New Connaught Place, Dehradun
India, p 2499
97. Mader TL, Brumm MC (1987) Effect of feeding Sarsaponin in cattle and swine diets. J Anim
Sci 65(1):9–15. https://doi.org/10.2527/jas1987.6519
98. Kahlon TS, Chapman MH, Smith GE (2007) In vitro binding of bile acids by okra, beets,
asparagus, eggplant, turnips, green beans, carrots, and cauliflower. Food Chem 103:676–680.
https://doi.org/10.1016/j.foodchem.2006.07.056
99. Wang NF, Zhang XJ, Wang SW, Guo QB, Li ZJ, Liu HH (2020) Structural characterisation
and immunomodulatory activity of polysaccharides from white asparagus skin. Carbohydr
Polym 227:115314. https://doi.org/10.1016/j.carbpol.2019.115314
100. Wang S, Zhu F (2016) Antidiabetic dietary materials and animal models. Food Res Int
85:315–331. https://doi.org/10.1016/j.foodres.2016.04.028
Therapeutic Properties of Herbal Constituents Subjected for Clinical Trials 1513
101. Zhao JJ, Zhao HJ, Zhao D, Wang JQ, Qu WJ (2012) Hypoglycemic activity of clarified
asparagus juice in streptozotocin induced diabetic rats. Nat Product Res Develop 24:460–464
102. Zhao HJ (2010) Study on functional components and biological activities of old stem extracts
from Asparagus officinalis L. East China Normal University, China
103. Hafizur RM, Kabir N, Chishti S (2012) Asparagus officinalis extract controls blood glucose
by improving insulin secretionβ-cell function in streptozotocin-induced type 2 diabetic rats.
Br J Nutr 108(9):1586–1595
104. Xu G, Kong W, Fang Z, Fan Y, Yin Y, Sullivan SA, Tran AQ, Clark LH, Sun W, Hao T,
Zhao L, Zhou C, Jump VL (2021) Asparagus officinalis exhibits anti-tumorigenic and anti-
metastatic effects in ovarian cancer. Front Oncol. https://doi.org/10.3389/fonc.2021.688461
105. Wang J, Zhao J, Liu Y, Pang X (2013) Saponins extracted from by-product of Asparagus offi-
cinalis L. suppress tumour cell migration and invasion through targeting rho GTPase signal-
ling pathway. J Sci Food Agric 93(6):1492–1498. https://doi.org/10.1002/jsfa.5922
106. Bousserouel S, Grandois JL, Gosse F, Werner D, Barth SW, Marchioni E, Marescaux J, Raul
F (2013) Methanolic extract of white asparagus shoots activates TRAIL apoptotic death path-
way in human cancer cells and inhibits colon carcinogenesis in a preclinical model. Int J
Oncol 43(2):394–404. https://doi.org/10.3892/ijo.2013.1976
107. Xiang J, Xiang Y, Lin S, Xin D, Liu X, Weng L, Chen T, Zhang M (2014) Anticamcer effects
of deproteinized asparagus polysaccharide on hepatocellular carcinoma in vitro and in vivo.
Tumour Biol 35(4):3517–3524. https://doi.org/10.1007/s13277-013-1464-x
108. Cheng W, Cheng Z, Xing D, Zhang M (2019) Asparagus polysaccharide suppresses the
migration, invasion, and angiogenesis of hepatocellular carcinoma cells partly by targeting
the HIF-1 alpha/VEGF signalling pathway in vitro. Evid Based Complement Alternat Med.
https://doi.org/10.1155/2019/3769879
109. Poormoosavi SM, Najafzadehvarzi H, Behmanesh MA, Amirgholami R (2018) Protective
effects of Asparagus officinalis extract against Bisphenol A- induced toxicity in Wistar rats.
Toxiol Rep 5:427–433. https://doi.org/10.1016/j.toxrep.2018.02.010
110. Ku YG, Bae JH, Ayala M, Vearasilp S, Namiesnik J, Pasko P (2017) Efficient three-
dimensional fluorescence measurements for characterization of binding properties in some
plants. Sensors Actuators B Chem 248:777–778. https://doi.org/10.1016/j.snb.2017.04.050
111. Middelbeek RJW, Chambers MA, Tantiwong P, Treebak JT, An D, Hirshman MF, Musi N,
Goodyear LJ (2013) Insulin stimulation regulates AS160 and TBC1D1 phosphorylation sites
in human skeletal muscle. Nutrition Diabetes 3:6–9. https://doi.org/10.1038/nutd.2013.13
112. Li J ( 2011) Study on extraction, purification, and lipid lower in related activity of old stem
polysaccharide from Asparagus officinalis L. East China Normal University, China
113. Chrubasik S, Droste C, Dragano N, Glimm E, Black A (2006) Effectiveness and tolerability
of the herbal mixture asparagus P on blood pressure in treatment-requiring antihypertensives.
Phytomedicine 13:740–742. https://doi.org/10.1016/j.phymed.2006.01.009
114. Huang YX (2017) Effect of instant asparagus powder on sleeping improvement. J Anhui
Agric Sci 45(11):80–82
115. Jager AK, Mohoto SP, Heerden FRV, Viljoen AM (2005) Activity of a traditional south
African epilepsy remedy in the GABA-benzodiazepine receptor assay. J Ethnopharmacol
96:603–606. https://doi.org/10.1016/j.jep.2004.10.005
116. Miura T, Yasueda A, Sakaue M, Maeda K, Hayashi N, Ohno S (2016) SUN-LB271: a double-
blind randomized controlled trial regarding the safety and efficacy of enzyme-treated aspara-
gus extract intake in healthy human subjects. Clin Nutr 35:S145. https://doi.org/10.1016/
S0261-5614(16)30627-6
117. Chwedorzewska KJ, Galera H, Kosiński I (2008) Plantations of Convallaria majalis L. as a
threat to the natural stands of the species: genetic variability of the cultivated plants and natu-
ral populations. Biol Conserv 141:2619–2624. https://doi.org/10.1016/j.biocon.2008.07.025
118. Erdogan OI, Gokbulut A (2017) Adonis sp., Convallaria sp., Strophanthus sp., Thevetia sp.,
and Leonurus sp.—cardiotonic plants with known traditional use and a few preclinical and
clinical studies. Curr Pharm Des 23:1051–1059
1514 E. Vatsa and M. Aggarwal
119. Higano T, Kuroda M, Jitsuno M, Mimaki Y (2007) Polyhydroxylated steroidal saponins from
the rhizomes of Convallaria majalis. Nat Prod Commun 2:531–536. https://doi.org/10.117
7/1934578X0700200504
120. Higano T, Kuroda M, Sakagami H, Mimaki Y (2007) Convallasaponin A, a new
5β-spirostanoltriglycoside from the rhizomes of Convallaria majalis. Chem Pharm Bull
55:337–339. https://doi.org/10.1248/cpb.55.337
121. Stansbury J, Saunders P, Winston D, Zampieron ER (2012) The use of convallaria and
Crataegus in the treatment of cardiac dysfunction. J Restro Med 1(1):107–111. https://doi.
org/10.14200/jrm.2012.1.1012
122. Morimoto M, Tatsumi K, Yuui K, Terazawa I, Kudo R, Kasuda S (2021) Convallotoxin, the
primary cardiac glycoside in lily of the valley induces tissue factor expression in endothelial
cells. Vet Med Sci 7(6):2440–2444. https://doi.org/10.1002/vms3.614
123. Khundmiri SJ (2014) Advances in understanding the role of cardiac glycosides in control of
sodium transport in renal tubules. J Endocrinol 222(1):R11–R24
124. Jelliffe RW (2014) The role of digitalis pharmacokinetics in converting atrial fibrillation and
flutter to regular sinus rhythm. Clin Pharmacokinet 53(5):397–407
125. Choi DH, Kang DG, Cui X (2006) The positive inotropic effect of the aqueous extract of
convallaria keiskeiin beating rabbit artia. Life Sci 79(12):1178–1185
126. Lehmann HD (1984) Effect of plant glycosides on resistance and capacitance vessels.
Arzneimittelforschung 34(4):423–429
127. Lateef T, Rukash H, Bibi F, Azmi M, Qureshi S (2010) Effect of convallaria majalison kid-
ney function. J Dow Univ Health Sci 4(3):94–97
128. Nartowska J, Sommer E, Pastewka K, Sommer S, Rozewska ES (2004) Anti-angiogenic
activity of convallamaroside, the steroidal saponin isolated from the rhizomes and roots of
Convallaria majalis L. Med Chem 61(4):279–282
129. Moxley RA, Schneider NR (1989) Apparent toxicosis associated with lily-of-the-valley
(Convallaria majalis) ingestion in a dog. J Am Vet Med Assoc 195:485–487
130. Fitzgerald KT (2010) Lily toxicity in the cat. Top Companion Anim Med 25(4):213–217.
https://doi.org/10.1053/j.tcam.2010.09.006
131. Launert E (1981) Covers plants in Europe. A drawing of each plant, quite a bit of interesting
information. Edible and Medicinal Plants. Hamlyn
Plant-Derived Immunomodulators
Targeting COVID-19 (SARS-CoV-2):
Preclinical Evaluation and Clinical Trials
Robin, Pardeep Kaur, Jagdeep Kaur, Kamaljit Kaur, and Sunidhi Miglani
1 Introduction
Robin
Agilent Technologies India Pvt. Ltd., Amritsar, Punjab, India
Regional Water Testing Laboratory, Department of Water Supply and Sanitation,
Government of Punjab, Amritsar, Punjab, India
P. Kaur (*)
Post Graduate Department of Botany, Khalsa College, Amritsar, Punjab, India
J. Kaur
Department of Botanical and Environmental Sciences, Guru Nanak Dev University,
Amritsar, Punjab, India
K. Kaur
Post Graduate Department of Biotechnology, Khalsa College, Amritsar, Punjab, India
S. Miglani
Department of Botany, Sikh National College, Banga, Punjab, India
needed. Several antiviral medications are being explored, including the nucleotide
analog remdesivir, although none has been explicitly allowed for COVID-19.
Treatment regime that addresses the immunopathology of infection has been a main
focus, in addition to the research and techniques in vaccines that directly target the
viral strain or inhibit entrance [4]. It has been shown that virus-specific T-cells are
accountable for the cell-mediated immunity, and B-lymphocytes are responsible for
the humoral immunity. Both play crucial roles in the adaptive immune response of
body in all pathogenic viral infections, including COVID-19. Neutrophils, interleu-
kin (IL)-6, and C-reactive protein are increased, but the overall number of lympho-
cytes decreases. T-lymphocyte activation contributes to the aggravation of
inflammatory reactions, while B-lymphocytes aid in the production of virus-specific
neutralizing antibodies. Immunomodulators act as vital stimulants to body for natu-
ral defense against pathogenic diseases including SARS-CoV-2. Immunopotentiating
herbal medicines are considered complementary to contemporary medicine; as a
result, natural immunomodulatory agents have been shown to be safe, nontoxic, and
effective substitutes. Phytoimmunomodulators boost the body’s natural defenses
against pathogens like viruses, allowing the immune system to maintain homeosta-
sis and feasibly preventing viral infections. Allergies, various cancers, inflammatory
bowel disease (IBD), autoimmune disorders, pathogenic viral infections, and a vari-
ety of other chronic illnesses are linked to immune system malfunctions or imbal-
ances. The use of plant-derived immunomodulators to control the present episode of
coronavirus infection may serve as a precedent, demonstrating that conventional
health care may contribute to the effective treatment of patients. The crucial har-
nessing of all knowledge systems accessible around the globe, on the other hand,
would be the desired course of action. The immunomodulatory properties of plants
have been extensively studied, and some of the most well-known are Ocimum sanc-
tum, Tinospora cordifolia, Withania somnifera, Glycyrrhiza glabra, and Emblica
officinalis, indicating that they have both preclinical and clinical potential against
SARS-CoV-2. The chapter addresses the beneficial effects of these phytoimmuno-
modulators in order to uncover further pieces of evidence in the fight against
COVID-19 via preclinical and clinical experiments.
2 Phytoimmunomodulators
Ocimum sanctum
2.1
Balkrishna et al. [10] reported the utilization of the humanized zebrafish model,
which was xeno-transplanted with the A549 lung epithelial human cells for assess-
ing the protective actions of the tri-herbal ayurvedic medicine (coronil) consisting
of O. sanctum (leaf extract) along with T. cordifolia and W. somnifera. The research-
ers presented immunomodulatory properties of this herbal combination in retrieving
the pathological features prompted by SARS-CoV-2 spike protein, proposing its
possible utilization in SARS-CoV-2 infection. The combination inhibited SARS-
CoV-2 spike protein, restored normal morphological and cellular characteristics in
the swim bladder, and attenuated the skin hemorrhage, degeneration of renal cells,
and necrosis. Besides this, herbal formulation attenuated IL-1, induced secretions of
IL-6 and tumor necrosis factor (TNF)-cytokine, and reduced the TNF-induced tran-
scriptional activity of nuclear factor-kappa B/activator protein 1 (NF-ĸB/AP-1) in
A549 cells. Bioactive molecules that might be responsible for this effect were iden-
tified by ultra high performance liquid chromatography (UHPLC), which included
ursolic acid, rosmarinic acid, withanone, withanoside IV–V, withaferin A, and other
compounds like cordifolioside A, magnoflorine, palmatine, and betulinic acid. In
further studies, the authors established that formulation containing O. sanctum as
one of the ingredients efficiently repressed ACE-2 interaction with wild-type S pro-
tein (SWT), high infectious variant (SD614G), and mutated variant (SW436R) with consid-
erably greater affinity toward ACE-2. It also prevented the cytokine response in
A549 cells mediated by SARS-CoV-2 S-protein pseudotyped vesicular stomatitis
virus (VSVppSARS-2S) by declining the entrance of the pseudoviruses to the host
cells [11].
Devpura et al. [12] designed their research study to assess the clinical efficacy of
O. sanctum and other potent medicinal plants in restraining the pathological symp-
toms of SARS-CoV-2 infection employing an ayurvedic treatment regime. The
researchers reported reduced fold changes in the serum levels of the high sensitive
C-reactive protein (hs-CRP), tumor necrosis factor alpha (TNF-α), and interleukin-
6 (IL-6), stimulating virological clearance and recovery and concomitantly reduc-
ing the viral dissemination risk.
Tinospora cordifolia
2.2
Withania somnifera
2.3
Glycyrrhiza glabra
2.4
[37]. In one of the studies, Gowda et al. [38] evaluated the efficacy of glycyrrhizin
as high-mobility group box 1 (HMGB1) inhibitor of SARS-CoV-2 viral protein
expression on the cellular perturbations in the lung cells. As severity of COVID-19
infection is associated with amplified inflammatory responses, HMGB1 (damage-
associated molecular pattern protein) is an imperative extracellular mediator in the
inflammatory process. It was observed that this natural triterpene of licorice inhib-
ited the viral protein-induced cell pyroptosis, and attenuated proinflammatory cyto-
kines IL-1β, IL-6, and IL-8 release as well as ferritin from the macrophages
cultivated in the lung cells expressing SARS-CoV-2 S-RBD. The researchers con-
cluded the dual capability of this compound to concurrently terminate viral replica-
tion and decline proinflammatory mediators, which could establish it as an appealing
therapeutic alternative in patients with SARS-CoV-2 infection.
Yu et al. [39] also found glycyrrhizin to be the most efficacious among various
active ingredients of traditional Chinese medicine and a nontoxic, broad-spectrum,
anti-coronavirus molecule, predominantly against SARS-CoV-2. Additionally, in
vitro experimental investigations conducted by Tolah et al. [40] also revealed robust
antiviral action of crude licorice extract and glycyrrhizin under various treatment
procedures (simulations of the treatment with the viral infection, post-infection
treatment, and the pre-treatment), signifying multifold mechanisms for its action.
The authors noted that half-maximal inhibitory concentration (IC50) of glycyrrhizin
was significantly lesser than licorice. Li et al. [41] provided experimental and simu-
lation data demonstrating high-affinity interaction of glycyrrhizin with the S-protein,
potentially targeting recombinant S-protein-mediated binding to the host cells
exhibiting its antiviral activity. It has also been observed that G. glabra extract
reduces the expression of ACE-2 at the protein level in the small bowel, which may
serve as an entrance point of SARS-CoV-2. The present findings supported the con-
cept that dietary supplementation of G. glabra extract might mitigate the accumula-
tion of viral strain in the gastrointestinal tract and therefore help in the avoidance of
possible fecal–oral SARS-CoV-2 spread [42]. Such studies highlight the primary
natural bioactives of G. glabra as prospective antiviral candidates that might be
further explored for the preventive treatment of COVID-19.
Derived from G. glabra roots, glycyrrhetinic acid is a hydrolytic product of glyc-
yrrhizic acid that functions as a glucocorticoid-like drug helping in reducing inflam-
mation and in immune regulation against cytokine storm. A case report of
nonhospitalized COVID-19 patients who failed to get relief with the regular treat-
ment of COVID-19 showed significant positive response toward glycyrrhizin as a
potential antiviral drug for the treatment of the disease. Although the patient recov-
ered from this viral illness, further controlled experiments are considered obligatory
to corroborate its therapeutic actions [43]. Later on, a research group in their single-
center observational study observed that the treatment accommodated immunologic
functions with lesser prevalence of new-onset impediments, which include the acute
respiratory distress syndrome, acute liver, and acute myocardial infarction [44]. An
open-label randomized controlled multicenter trial was implemented in Iran on hos-
pitalized adult patients employing herbal decoction and capsule containing G. gla-
bra as one of the components. Glycyrrhizin (29.505 μg/mL) was found to be the
Plant-Derived Immunomodulators Targeting COVID-19 (SARS-CoV-2): Preclinical… 1523
main marker of this plant via high performance liquid chromatography (HPLC)
analysis. The results of this study presented significant accelerated clinical improve-
ment of symptoms with decrease in muscle pain, chills, headache, dry cough,
fatigue, anorexia, sputum, runny nose, and vertigo in COVID-19 treatment group
[45]. One pilot, nonrandomized study (NCT04487964) is on-going considering the
extract of this plant as a complementary ayurvedic medicine in patients with SARS-
CoV-2 disease [46].
Emblica officinalis
2.5
Extract/
formulation
(concentration/ Isolated compounds/primary In vitro assay/cell culture Bioactivity/possible mechanism of
Plant dose) bioactive ingredients used/animal model action Reference
Ocimum Zebrafish model: Ursolic acid, betulinic acid, Humanized zebrafish model Inhibition of SARS-CoV-2 Balkrishna
sanctum, 12 and 58 μg/kg rosmarinic acid, cordifolioside A, (N = 24) spike-protein-induced renal cell et al. [10]
Tinospora A549:1–1000 μg/ magnoflorine, palmatine, A549 cells necrosis
cordifolia, and mL withanone, withaferin A, and Attenuation of the interleukin
Withania withanoside IV–V (IL)-1β induced IL-6 and TNF-α
somnifera cytokine secretions
Decrease in TNF-α induced NF-ĸB/
AP-1 transcriptional activity
O. sanctum, T. 0.001–30 μg/mL Rosmarinic acid, betulinic acid, A549 cells Inhibition of the interaction of Balkrishna
cordifolia, and ursolic acid, magnoflorine, ACE-2 with S proteins (SWT, SD614G, et al. [22]
W. somnifera palmatine, cordifolioside A, SW436R)
withaferin A, withanoside IV and Prevention of SARS-CoV-2
V, and withanone S-protein pseudotyped vesicular
stomatitis virus (VSVppSARS-2S)-
mediated cytokine response
T. cordifolia 6, 28, and 142 μg/ Cordifolioside A, magnoflorine, Humanized zebrafish model Reduction in number of granulocytes Balkrishna
kg/day β-ecdysone, and palmatine and macrophages et al. [20]
Restoration of behavioral fever, skin
hemorrhage, and mortality induced
by SARS-CoV-2 spike protein.
Robin et al.
Glycyrrhiza Ethyl acetate Liquiritin, 3,3′,4,4′-tetrahydroxy- Chromogenic assay Factor Xa inhibition Ibrahim
glabra fraction (1 mg/ 2-methoxychalcone, naringenin et al. [36]
mL) 5-O-glucoside, and
7-hydroxy-4′-methoxyisoflavone
Aqueous root Glycyrrhizic acid Vero E6 cells SARS-CoV-2 main protease (Mpro) van de
extract inhibition Sand et al.
(0.004–4 mg/mL) [37]
1 mM Glycyrrhizic acid SARS-CoV-2-S-RBD and Prevention of S-RBD- and Orf3a- Gowda
Orf3 transfected BEAS-2B mediated cell death and caspase-1 et al. [38]
cells; Vero E6 cells activation; dampening of
macrophage activation; attenuation
of proinflammatory cytokines IL-6
and CXCL18; diminution of ferritin
release; inhibition of SAR-CoV-2
replication
Glycyrrhizic acid Mouse aorta smooth muscle Inhibition against spike protein of Yu et al.
cells (MASMCs) and human SARS-CoV-2 [39]
bronchial epithelial (16HBE)
cells
Crude licorice Glycyrrhizin between 26.5 African green monkey kidney Antiviral activity against SARS- Tolah et al.
312.5 and 100 ng/ and 850 ng/mL cells Vero E6; human CoV-2 via the targeting of Mpro [40]
mL SARS-CoV-2 clinical patient
isolate (SARS CoV2/human/
SAU/85791C/2020)
Aqueous root Glycyrrhizinic acid Sprague-Dawley rats (N = 48, Reduction in ACE-2 mRNA levels Jezova
extract (150 mg/ male) and ACE-2 protein content in the et al. [42]
kg/day for small intestine
5 weeks)
Plant-Derived Immunomodulators Targeting COVID-19 (SARS-CoV-2): Preclinical…
ACE-2 angiotensin converting enzyme-2, BEAS-2B human bronchial epithelial cell line, CXCL18 C-X-C motif chemokine ligand 18, mRNA messenger ribo-
nucleic acid, NF-ĸB/AP-1 nuclear factor-kappa B/activator protein 1, SARS-CoV-2 Severe Acute Respiratory Syndrome Coronavirus-2, S-RBD spike protein
receptor-binding domain, TNF-α tumor necrosis factor-alpha
1527
1528 Robin et al.
Overall, Tables 1 and 2 summarize the potential in vivo and in vitro beneficial
effects of Ocimum sanctum, Tinospora cordifolia, Withania somnifera, Glycyrrhiza
glabra, and Emblica officinalis against COVID-19. Due to their bioaction in numer-
ous steps of the viral infection, such as inhibiting transmembrane protease serine 2
(TMPRSS2) and disrupting viral entry via binding to viral fusion proteins and
blocking angiotensin-converting enzyme-2 (ACE-2) receptor, these immunopoten-
tiators as well as their constituents may well be considered as potential adjuvant
therapies to COVID-19 in clinics. By revitalizing the human immune system, plant-
derived bioactive immunomodulators offer sustenance to immunity and aid in the
fight against SARS-CoV-2.
3 Conclusion
A variety of plant extracts, herbal medicines, and pure plant ingredients have been
demonstrated to have immunomodulatory properties and have been utilized as pre-
ventative or therapeutic treatments for infectious disorders. Through the activation
and modulation of the lymphocytes, macrophages, and cytokine production, these
phytocomponents and their bioactive chemicals influence immunological responses.
Many plant extracts include a variety of bioactive components, such as polysaccha-
rides, alkaloids, glycosides, terpenoids, flavonoids, and essential oils, all of which
have a powerful capacity to sustain and/or activate the immune system via modula-
tion of nonspecific immunological responses. The current analysis emphasizes the
relevance of medicinal plants that contain immunomodulating components of vari-
ous chemistries that might be used to combat viral infections. Therefore, research-
ers’ attempts to create alternative plant-based medicines for infectious illnesses
should be encouraged.
References
1. Nakamura T, Isoda N, Sakoda Y, Harashima H (2022) Strategies for fighting pandemic virus
infections: integration of virology and drug delivery. J Control Release 343:361–378
2. Yuki K, Fujiogi M, Koutsogiannaki S (2020) COVID-19 pathophysiology: a review. Clin
Immunol 215:108427. https://doi.org/10.1016/j.clim.2020.108427
3. Nalbandian A, Sehgal K, Gupta A et al (2021) Post-acute COVID-19 syndrome. Nat Med
27:601–615. https://doi.org/10.1038/s41591-021-01283-z
4. Cao X (2020) COVID-19: immunopathology and its implications for therapy. Nat Rev
Immunol 20:269–270. https://doi.org/10.1038/s41577-020-0308-3
5. Kaur P, Robin MVO, Arora R, Singh B, Arora S (2017) Immunopotentiating significance of
conventionally used plant adaptogens as modulators in biochemical and molecular signal-
ling pathways in cell mediated processes. Biomed Pharmacother 95:1815–1829. https://doi.
org/10.1016/j.biopha.2017.09.081
6. Varshney KK, Varshney M, Nath B (2020) Molecular modeling of isolated phytochemicals
from Ocimum sanctum towards exploring potential inhibitors of SARS coronavirus main
Plant-Derived Immunomodulators Targeting COVID-19 (SARS-CoV-2): Preclinical… 1529
39. Yu S, Zhu Y, Xu J et al (2021) Glycyrrhizic acid exerts inhibitory activity against the spike protein
of SARS-CoV-2. Phytomedicine 85:153364. https://doi.org/10.1016/j.phymed.2020.153364
40. Tolah AM, Altayeb LM, Alandijany TA et al (2021) Computational and in vitro experimental
investigations reveal anti-viral activity of licorice and glycyrrhizin against severe acute respira-
tory syndrome coronavirus 2. Pharmaceuticals (Basel) 14(12):1216. https://doi.org/10.3390/
ph14121216
41. Li J, Xu D, Wang L et al (2021) Glycyrrhizic acid inhibits SARS-CoV-2 infection by blocking
spike protein-mediated cell attachment. Molecules 26(20):6090
42. Jezova D, Karailiev P, Karailievova L et al (2021) Food Enrichment with Glycyrrhiza Glabra
Extract Suppresses ACE2 mRNA and Protein Expression in Rats–Possible Implications for
COVID-19. Nutrients 13(7):2321. https://doi.org/10.3390/nu13072321
43. Ding H, Deng W, Ding L et al (2020) Glycyrrhetinic acid and its derivatives as potential alter-
native medicine to relieve symptoms in nonhospitalized COVID-19 patients. J Med Virol
92(10):2200–2204
44. Tan R, Xiang X, Chen W et al (2021) Efficacy of diammonium glycyrrhizinate combined with
vitamin C for treating hospitalized COVID-19 patients: a retrospective, observational study.
QJM Int J Med hcab184. https://doi.org/10.1093/qjmed/hcab184
45. Karimi M, Zarei A, Soleymani S et al (2021) Efficacy of Persian medicine herbal formulations
(capsules and decoction) compared to standard care in patients with COVID-19, a multicenter
open-labeled, randomized, controlled clinical trial. Phytother Res 35(11):6295–6309
46. Gomaa AA (2020) Complementary intervention for COVID-19 (Clinicaltrials.gov Identifier:
NCT04487964). Retrieved from https://clinicaltrials.gov/ct2/show/NCT04487964
47. Chikhale RV, Sinha SK, Khanal P et al (2021) Computational and network pharmacology stud-
ies of Phyllanthus emblica to tackle SARS-CoV-2. Phytomedicine Plus 1(3):100095
48. Murugesan S, Kottekad S, Crasta I et al (2021) Targeting COVID-19 (SARS-CoV-2) main
protease through active phytocompounds of ayurvedic medicinal plants–Emblica officinalis
(Amla), Phyllanthus niruri Linn.(Bhumi Amla) and Tinospora cordifolia (Giloy)–a molecular
docking and simulation study. Comput Biol Med 136:104683
49. Wu C, Liu Y, Yang Y et al (2020) Analysis of therapeutic targets for SARS-CoV-2 and discov-
ery of potential drugs by computational methods. Acta Pharm Sin B 10(5):766–788
50. Rangnekar H, Patankar S, Suryawanshi K et al (2020) Safety and efficacy of herbal extracts
to restore respiratory health and improve innate immunity in COVID-19 positive patients with
mild to moderate severity: a structured summary of a study protocol for a randomised con-
trolled trial. Trials 21:943. https://doi.org/10.1186/s13063-020-04906-x
Correction to: Bioprospecting
of Tropical Medicinal Plants
Correction to:
Chapters 4 and 5 in: K. Arunachalam et al. (eds.),
Bioprospecting of Tropical Medicinal Plants,
https://doi.org/10.1007/978-3-031-28780-0
© The Editor(s) (if applicable) and The Author(s), under exclusive license to 1533
Springer Nature Switzerland AG 2023
K. Arunachalam et al. (eds.), Bioprospecting of Tropical Medicinal Plants,
https://doi.org/10.1007/978-3-031-28780-0
1534 Index
Antibacterial, 9, 13, 14, 112, 120, 180, 189, 1256–1258, 1329, 1331, 1335, 1336,
195, 196, 200, 216, 217, 257, 271, 278, 1339, 1351, 1353, 1368, 1394, 1413,
316, 395, 411–413, 415, 419, 484, 487, 1420, 1421, 1461, 1463, 1465–1467,
489, 514, 518, 548, 555, 590, 591, 1487, 1497, 1499–1502, 1504, 1517,
594–596, 601, 605, 606, 650, 678, 679, 1518, 1525
686, 707, 714, 722, 735, 809, 810, Antimicrobial, 5, 6, 11, 12, 19, 116, 120, 172,
815–816, 961, 974, 980, 1050, 1065, 178, 200, 208, 216, 271, 272, 278, 374,
1067, 1068, 1100, 1105, 1118, 1212, 395, 410, 412–415, 419, 421, 429, 447,
1267, 1268, 1270, 1271, 1273, 1277, 465, 469, 484, 485, 490, 511, 518, 529,
1280, 1349, 1353, 1358, 1465, 1467, 532, 535, 546, 553–555, 589, 590, 595,
1468, 1517 604–606, 612, 650, 656, 662–680, 686,
Antibacterial activity, 361, 395–398, 469, 553, 700, 727, 735, 736, 749, 753, 790, 793,
554, 592, 595, 596, 600, 601, 604–607, 798–799, 809–810, 813–817, 864, 894,
677, 678, 680, 722, 813–814, 900, 933, 935, 937, 962, 978, 979, 1040,
1127, 1270, 1273–1276, 1278–1281, 1048, 1049, 1051, 1064, 1065, 1067,
1358, 1359 1106, 1125–1127, 1130, 1251, 1252,
Antibiotics, 116, 267, 274, 278, 415, 421, 482, 1265–1282, 1336, 1347–1353, 1357,
604, 605, 815, 912, 915, 916, 1007, 1358, 1360, 1465–1468, 1499, 1502
1008, 1050, 1067, 1266–1271, 1281, Antimicrobial agents, 413, 415, 469–470, 606,
1348, 1351, 1352, 1357, 1360, 1067, 1282, 1347–1360
1435, 1499 Antimicrobial synergism, 1270
Anticancer, 8, 12–14, 16, 18, 20, 175, 178, Antioxidant, 5, 6, 10, 120, 172, 195, 200, 216,
200, 257, 267, 271, 274, 278, 279, 296, 261, 263, 266, 267, 269–274, 278, 296,
395, 399–400, 411–413, 418, 419, 421, 364, 366, 368, 372, 374, 395, 401,
427–450, 466, 470, 489, 490, 495, 502, 410–413, 418, 419, 421, 427, 429, 443,
513, 514, 518, 538, 546, 547, 554, 555, 465, 467, 469, 473, 484–487, 490, 494,
565–581, 591, 636–638, 650, 683–685, 496–497, 500–502, 511, 513, 518, 529,
700, 727–736, 778, 779, 790, 797–799, 533–535, 548, 550–555, 589–592, 594,
894, 961, 979, 980, 1040, 1041, 1050, 595, 599–607, 619, 634, 636, 640, 642,
1065, 1066, 1097, 1105, 1118, 650, 680–683, 686, 704, 705, 707, 727,
1127–1130, 1136, 1137, 1140–1159, 735, 736, 745, 747, 749, 753, 774, 778,
1161–1163, 1178, 1191, 1336, 1339, 779, 790, 792, 797, 799, 807, 816, 817,
1353, 1368, 1369, 1374, 1379, 1408, 864, 871, 879, 880, 894, 900, 928, 929,
1413, 1417, 1418, 1422, 1465–1468, 937, 938, 943, 944, 951, 955, 961, 962,
1497, 1504 965–967, 973–980, 983, 987, 1040,
Anticancer agents, 279, 403, 470, 512, 567, 1050, 1052, 1067, 1070, 1097, 1098,
569, 572, 779, 1161, 1193, 1369, 1449 1100–1102, 1104–1107, 1118, 1119,
Antiepileptic drugs (AEDs), 821–858 1121, 1122, 1125–1130, 1163, 1212,
Anti-inflammatory, 5, 7, 9, 13, 14, 111–118, 1214, 1216, 1217, 1240, 1250–1252,
120, 126, 127, 130, 134, 139, 150, 151, 1256, 1279, 1329, 1331, 1332,
172, 175, 178, 180, 184, 187, 189, 194, 1336–1340, 1348, 1353, 1360, 1368,
216, 263, 265–267, 269–274, 278, 279, 1393, 1407, 1420, 1422, 1423,
294, 297, 300, 367–372, 374, 382, 395, 1465–1467, 1496, 1497, 1499, 1500,
400–401, 410–412, 419–421, 429, 1502, 1504, 1518
447–449, 483, 484, 487, 490, 497, Antiparasitic, 413, 567, 1052, 1064,
499–502, 518, 532, 533, 546, 551, 555, 1118, 1467
567, 590, 591, 603, 639, 650, 683–686, Antiproliferation, 554
735, 736, 744, 745, 747, 749, 753–757, Apocynaceae, 20, 60, 75, 88, 90, 110, 112,
778, 790, 791, 798, 799, 869, 894, 961, 117, 118, 125, 131, 132, 136, 139, 140,
962, 974, 975, 979, 980, 983, 1041, 145, 147, 148, 151, 153, 231, 233, 248,
1050, 1052, 1065–1068, 1097, 1099, 341, 343, 349, 368–369, 428, 484, 511,
1101, 1104–1107, 1118, 1125, 778, 835, 846, 1017, 1018, 1021, 1062,
1129–1130, 1199, 1200, 1203–1221, 1063, 1140, 1141, 1144, 1150, 1193,
1236, 1239, 1240, 1251–1254, 1194, 1314, 1472
Index 1535
Apoptosis, 412, 432–449, 502, 534–538, 554, 1265, 1266, 1271, 1281, 1330, 1418,
555, 569–573, 575–579, 613–615, 619, 1448, 1450, 1495, 1497, 1519,
636, 639, 797, 799, 814, 870, 879, 962, 1521, 1523
975, 1066, 1098, 1118, 1128, 1129, Bioactives, 58, 91, 255, 257, 269, 270, 272,
1144, 1146–1148, 1150, 1152–1154, 294–300, 348, 352, 372, 373, 390, 395,
1157, 1158, 1160, 1161, 1187, 403, 411, 413–415, 418–420, 430,
1191–1193, 1254, 1331, 1339, 1366, 432–442, 450, 466–469, 511, 517–518,
1369, 1370, 1372, 1375, 1376, 1395, 522, 530, 547, 565, 568–570, 574, 603,
1405–1407, 1412, 1413, 1418, 1419, 606, 634, 636, 650, 679, 686, 709, 725,
1422, 1449, 1500–1502 727, 728, 731, 734–736, 739–757, 773,
Asparagus, 5, 20, 21, 58, 62, 111, 126, 129, 209, 774, 776–778, 791, 799, 817, 858, 872,
262, 267, 274, 279, 382, 603, 869, 981, 929, 938, 939, 971–989, 1047–1049,
1141, 1160, 1305, 1503–1505, 1523 1051, 1052, 1056–1058, 1066, 1101,
Assam, 2, 227–229, 231–233, 239, 300, 1118, 1119, 1186, 1199, 1200, 1206,
659–661, 664, 672–676 1208, 1216, 1236–1238, 1248, 1250,
Astaxanthin, 1051 1254, 1256, 1265, 1266, 1269, 1271,
Asthma, 4, 8, 9, 12–14, 16, 18, 21, 55, 60, 68, 1272, 1278, 1281, 1330, 1332, 1413,
72, 74, 77, 81, 83, 86, 88, 89, 101, 111, 1418–1420, 1448, 1450, 1485, 1495,
114, 116, 117, 146, 149, 173, 175–177, 1497, 1502, 1504, 1517–1528
179, 185, 192, 193, 196, 198, 200, 206, Bioactivity, 269, 313, 403, 449, 469, 545,
208, 209, 211, 214, 217, 228, 229, 550–557, 570, 736, 744, 749, 791, 855,
231–233, 239, 263, 266, 268, 274, 297, 938, 943, 1066, 1118, 1119, 1238,
336, 339, 340, 342, 352, 471, 483, 489, 1405, 1407, 1526
498, 501, 790, 798, 979, 983, 1038, Biochanin A, 493, 789–800
1121, 1199, 1200, 1203, 1209, 1353, Biocompatibility, 1370, 1372, 1375, 1389,
1357, 1460, 1461, 1472 1391, 1393, 1399, 1410, 1414, 1418,
Atherosclerotic plaques, 867, 876 1422, 1498
Attappady, 107–120 Biodiversity, 2, 159, 160, 163, 292, 310, 312,
Ayurveda, 1, 123, 227, 254–260, 264, 316–321, 323, 330, 357, 373, 701, 767,
269–273, 279, 281, 287–289, 292, 307, 781–783, 785, 789, 1005, 1053,
429, 489, 545, 612, 649, 768, 927, 928, 1088, 1486
932, 933, 935, 1042, 1103, 1119, 1121, Bioprospecting, 25, 312, 317–318, 322–324
1299–1319, 1329, 1332, 1470, Brain functions, 822, 852
1518, 1523 Brassinosteroids, 1048
Ayurvedic medicines, 255, 258, 281, 295, 487,
488, 500, 707, 932, 1127, 1518,
1519, 1523 C
Azadirachta indica, 58, 62, 94, 96, 99, 110, Cancer, 7, 13, 14, 18, 55, 112, 128, 186, 196,
111, 126, 129, 231, 238, 262, 265, 200, 205, 209, 210, 228, 244, 245, 247,
1251–1252, 1314 266–269, 296, 318, 342, 387, 400–403,
418, 419, 427–450, 470, 483, 495, 501,
502, 511, 512, 519, 520, 522, 529–540,
B 550, 555, 556, 565–567, 569–580, 590,
β-amyloid plaques, 273, 1329 604, 611, 612, 616–620, 636, 638, 640,
Bioactive components, 482, 530, 531, 642, 643, 686, 744, 747, 768, 778–782,
537–539, 555, 571, 606, 744, 776, 790, 791, 794, 797, 798, 809, 812, 815,
984–985, 1047, 1104, 1106, 1528 816, 863, 868, 870, 876, 938, 974–977,
Bioactive compounds, 58, 269, 270, 272, 294, 1042, 1050, 1051, 1065, 1097, 1099,
295, 297, 300, 373, 403, 411, 414, 415, 1118, 1125, 1127–1129, 1136–1164,
530, 547, 565, 574, 650, 679, 686, 727, 1177–1194, 1199, 1216, 1248, 1251,
735, 736, 739–757, 773, 774, 777, 1302, 1335, 1357, 1359, 1365–1378,
971–989, 1047–1049, 1052, 1387–1401, 1405–1423, 1429–1450,
1056–1057, 1118, 1199, 1200, 1206, 1471, 1474, 1480, 1481, 1485, 1497,
1208, 1236–1238, 1248, 1256, 1499, 1501, 1504, 1516
1536 Index
Cancer cell lines, 361, 370, 373, 399, 400, 1495, 1496, 1498, 1502, 1503,
430–436, 438–450, 470, 495, 501, 535, 1505–1507, 1520, 1521, 1524, 1525
554, 555, 568, 570–580, 685, 1066, Clustered often interspaced short palindromic
1067, 1119, 1127–1129, 1160, 1189, repeats (CRISPR), 1054, 1055
1192, 1194, 1372–1374, 1376, 1412, Cognitive decline, 1237, 1327
1413, 1417, 1420, 1422 Computer-aided drug design (CADD),
Cancer therapeutics, 501, 1137, 1138 855, 857
Cancer treatment, 428, 448, 565, 568, 574, Conservation, 21, 22, 47, 50, 102, 160, 163,
611, 745, 778, 1161, 1164, 1189, 1190, 167, 222, 223, 287, 291, 314–324, 330,
1194, 1365, 1367, 1368, 1388–1396, 333, 353, 382, 385, 404, 410, 421, 783,
1399, 1423, 1448 1006, 1012–1021, 1029, 1035–1037,
Carbon nanotubes (CNTs), 1179, 1368–1371, 1039, 1040, 1042, 1043, 1062, 1067,
1379, 1421 1082, 1088–1089, 1486
Cardiovascular disease (CVD), 52, 97, 98, Conservation and utilization, 1088
269, 427, 518, 522, 863, 864, 866–872, Conservation measures, 1036
876–880, 975, 977, 987, 989, 1097, Cosmo-ceuticals, 464, 472–473
1216, 1217, 1429, 1497 Cost effectiveness, 1448, 1487, 1488
Catharanthus, 6, 20, 21, 112, 132, 248, 318, COVID-19, 799, 1266, 1319, 1360,
428, 482, 511–523, 769, 778, 779, 1515–1528
1063, 1065, 1141, 1194, 1368, 1407 CRISPR-associated9 (Cas9)
Cell signalling, 811, 880, 1119 endonuclease, 1054
Characterization, 403, 516, 639, 641, 642, Cryotherapy, 501, 1057
712–713, 771–778, 858, 1055, 1177, Culture, 1, 21, 25, 44, 102, 107, 108, 167, 227,
1184–1186, 1237, 1397, 1495 244, 249–251, 254, 288, 289, 316, 320,
Chemical carcinogens, 1439 347, 360, 374, 385, 466, 482, 486, 516,
Chemotherapeutics, 2, 427, 428, 430, 445, 555, 596, 600, 601, 662, 707–709,
450, 512, 568, 572, 573, 611, 614, 615, 714–716, 723, 725, 880, 910, 912, 920,
618, 620, 826, 1129, 1177, 1189, 1365, 921, 982, 1006, 1010, 1011, 1022,
1366, 1413, 1422, 1499 1024, 1026, 1027, 1029, 1035,
Chemotherapy, 427, 470, 514, 518–520, 565, 1039–1042, 1049, 1055–1058, 1061,
574, 611–620, 798, 870, 983, 1057, 1081–1090, 1100, 1107, 1395, 1422,
1099, 1118, 1160–1162, 1193, 1194, 1480, 1489, 1521, 1526
1367, 1387–1401, 1408, 1442, 1447 Curcumin, 278, 428, 436, 445, 447, 448, 484,
Chittur, 123–156 590, 637, 779, 810, 814, 928, 961,
Chromatographic analysis, 967 1058, 1097–1100, 1138, 1152, 1206,
Cinnamomum, 113, 132, 203, 231, 236, 263, 1208, 1218, 1235, 1238–1240, 1254,
268, 313, 649–686, 1142, 1259, 1279, 1312, 1369, 1371, 1372,
1273, 1314 1374–1378, 1423, 1449
Cinnamomum cassia, 650, 657, 658, 661, 662, Current scenario, 1328
680–684, 686, 1253, 1271, 1339 Cuttings propagation, 1086
Cinnamon, 662, 678, 679, 681, 682, 684, 686, Cyclea peltata, 114, 703, 707–725
1103–1105, 1237, 1269, 1273, 1339, Cyclooxygenase-1 (COX-1), 869, 879, 1068,
1352, 1353, 1358, 1496 1130, 1203, 1204, 1209, 1211
Climber, 27, 58, 90, 119, 125, 172–174, Cytotoxic effect, 362, 418, 432, 434, 437, 444,
178–180, 182, 185–187, 192, 194, 196, 446, 447, 449, 556, 567, 570, 573, 577,
202, 204, 209, 210, 215, 233, 339, 363, 685, 1129, 1160, 1338, 1374, 1375
364, 703, 1036, 1251, 1470
Clinical status, 1377, 1378
Clinical trials, 254, 270, 279, 281, 403, 411, D
482, 501, 520, 611, 617, 638, 778, 791, Data analysis, 44, 51–57, 334
800, 857, 865, 1097–1107, 1177, 1187, Dendrimers, 1368, 1373, 1374, 1379, 1421
1190, 1194, 1328, 1377, 1378, 1401, Desert, 270, 297, 330–345, 347–349,
1405, 1406, 1410, 1412, 1416, 1420, 351–353, 1005, 1062
1438, 1449, 1475, 1483, 1484, 1489, Dhemaji, 228, 229, 231–233
Index 1537
Diabetes, 6, 8, 10, 11, 13, 15, 18, 53, 71, 1373–1376, 1388, 1391–1392, 1396,
73–75, 79–81, 84, 85, 88, 98, 100, 115, 1398–1401, 1410, 1415, 1421
116, 118, 126, 137, 143, 152, 179, 180, Drug discovery, 102, 280, 281, 351, 372–374,
186, 188, 190, 195, 199, 209, 217, 382, 409, 411, 416, 421, 481, 488, 553,
245–247, 262, 265, 266, 268, 269, 274, 557, 649, 767–785, 789, 858, 929,
279, 299, 343, 411, 412, 466, 470, 471, 1212, 1495, 1506
485–487, 489, 494, 497, 501, 502, 511,
552, 553, 638, 640, 650, 743, 745, 757,
794, 863, 866–868, 944, 965, 974, 975, E
977–979, 983, 989, 1038, 1040, 1097, Emblica officinalis, 261, 263, 265, 1376, 1516,
1098, 1101, 1104–1107, 1125, 1199, 1523–1528
1247, 1249–1252, 1260, 1303, 1406, Endangered plants, 1029, 1039–1041,
1496–1499 1081–1083, 1087–1089
Dietary supplement, 638, 640, 753, 1056, Endothelial NOS (eNOS), 799, 879
1414, 1481, 1482 Enhancing cancer therapy, 1413
Diseases, 1, 3, 4, 12, 17, 20, 21, 27, 40, 45, 51, Environmental factors, 222, 1057, 1136, 1201,
52, 54, 55, 67, 72, 80, 89, 92, 93, 1328, 1438–1440, 1443, 1446, 1447
96–98, 100–102, 109, 118–120, 123, Enzymes, 269, 273, 274, 401, 402, 447, 448,
124, 126, 130, 132, 150, 152, 154, 155, 468, 497, 512, 516, 533, 547, 551–555,
174–177, 179, 182, 184–186, 189, 190, 604, 624, 626–629, 631–634, 641, 715,
193, 195–197, 200, 202, 203, 206, 207, 747, 753, 756, 791, 794, 797, 809, 813,
215, 216, 227, 228, 230–233, 239, 244, 816, 817, 826, 854, 856, 865, 869, 871,
245, 253, 255–262, 269, 271–273, 275, 906, 908, 918–923, 943, 945, 962, 973,
278, 281, 289, 290, 292, 298, 309, 314, 976, 978, 980, 982, 1022, 1048, 1051,
349–352, 360–372, 374, 386, 387, 400, 1055, 1058, 1064, 1068, 1098–1101,
401, 403, 410, 411, 414, 415, 420, 448, 1105–1107, 1137, 1139, 1144, 1190,
464, 466, 469, 482–485, 487, 489, 490, 1191, 1193, 1203, 1213, 1217, 1238,
494, 496, 502, 511, 519, 522, 529, 533, 1240, 1258, 1332, 1334, 1336–1338,
534, 545, 546, 550, 551, 554, 557, 565, 1349, 1351, 1352, 1413, 1417, 1418,
604, 624, 629, 631, 638–640, 643, 707, 1442, 1443, 1482, 1496, 1498, 1505,
728, 735, 739, 745, 747, 769, 780, 783, 1517, 1521
790, 791, 796, 798, 799, 814, 822, 823, Epigenetics, 448, 1136–1164, 1442, 1443,
825–827, 857, 863–880, 905, 906, 916, 1447, 1449
917, 927–929, 932, 933, 935, 938, 939, Epilepsy, 7, 17, 115, 117, 120, 173, 188, 199,
941, 944, 961, 965, 971–989, 1035, 217, 273, 298, 412, 414, 418, 701, 704,
1036, 1038, 1039, 1042, 1043, 1050, 821–827, 829–831, 847, 850, 851, 853,
1051, 1057, 1061, 1062, 1064, 1065, 854, 858, 933–935, 1038, 1300,
1067, 1097–1099, 1106, 1107, 1505, 1506
1117–1119, 1121, 1125, 1127, 1129, Essential oils (EOs), 18, 176, 179, 186, 198,
1136, 1139, 1160–1162, 1164, 1194, 208, 212, 272, 296, 297, 363, 364, 366,
1199–1203, 1205–1207, 1209, 1212, 413–418, 421, 428, 430, 434, 436–438,
1217, 1233–1242, 1247–1252, 442, 444, 446, 447, 450, 546, 554, 556,
1254–1260, 1268, 1270, 1275, 567, 569, 572, 612, 649–651, 654,
1300–1304, 1314, 1318, 1327, 656–665, 672, 675, 677–680, 682–686,
1330–1332, 1334–1338, 1340, 1341, 753, 837, 840, 841, 843, 893–900,
1348, 1353, 1357, 1359, 1360, 1365, 933–938, 943, 944, 952, 961, 965,
1371, 1406, 1412, 1419, 1429, 1430, 1049, 1050, 1062–1064, 1067,
1433–1435, 1438, 1441–1445, 1266–1269, 1273–1276, 1278, 1279,
1447–1449, 1457, 1460, 1464, 1468, 1304, 1316–1318, 1336, 1347, 1348,
1471, 1479–1481, 1483, 1484, 1486, 1351–1353, 1357–1359, 1504,
1488, 1489, 1497, 1502, 1507, 1515, 1506, 1528
1516, 1519, 1522, 1523 Ethnobotanical, 1–22, 25, 43–103, 108–110,
Drug delivery, 619, 748, 1163, 1164, 124, 126, 239, 289, 333–334, 358, 410,
1177–1194, 1255, 1366, 1370, 411, 416, 466, 486, 488–490, 1118
1538 Index
Ethno medicinal plant, 171–222 512, 532, 547, 589–607, 617, 623–629,
Ethnomedicine, 1, 40, 227, 228, 290, 296, 333, 631–635, 637–640, 649, 662, 679, 701,
352, 360–372, 382, 383, 428, 429, 768 704, 727, 740–744, 747, 753, 756, 757,
Ethnopharmacology, 50, 318 782, 790, 807–817, 863–865, 880, 893,
Exosomes, 1087, 1375, 1379 894, 905, 917, 928, 935, 944, 965, 966,
Explants sterilization, 716, 1007 971–973, 977–989, 1047, 1055–1057,
Extract, 5–15, 17, 19–21, 40, 57, 126–153, 1061, 1063, 1071, 1097, 1117, 1125,
172, 174–177, 181, 183, 186–192, 1189, 1234–1237, 1241, 1265–1267,
194–197, 200, 202, 206–209, 216, 217, 1269, 1271, 1272, 1274–1277, 1279,
234–239, 258, 261–263, 269, 271–274, 1281, 1304, 1335, 1348, 1357, 1358,
280, 299, 313, 322, 343, 345, 350, 361, 1388, 1414, 1417, 1423, 1439, 1443,
363, 366–369, 371, 373, 388, 390, 395, 1445, 1446, 1449, 1457, 1482,
397, 399–403, 411, 414–416, 418, 420, 1505, 1517
428, 430–446, 448–450, 467–471, 473, Food microbiota, 807, 808, 817
482, 484, 487, 489, 494–501, 511, 516, Food toxicants, 1429, 1443
518, 520–523, 530–536, 538, 546–548, Fourier transformed infrared spectroscopy
551–557, 567–575, 591–606, 615, 617, (FTIR), 592, 594, 599, 602, 713,
629, 634, 651, 655, 657, 659, 663–665, 719–722, 775, 1416
668, 670, 674, 677–686, 700, 703, Fruits, 7, 12–16, 19, 20, 45, 58, 90, 125–134,
707–715, 717–722, 725, 728–736, 745, 136–138, 142, 144–146, 148, 150, 151,
746, 770, 771, 773–780, 782, 797, 809, 153, 173, 174, 177, 178, 181, 182, 184,
839, 841, 842, 844, 845, 847, 865, 187, 192, 193, 196, 203–205, 230,
869–871, 877, 930, 933, 936, 938–945, 234–239, 243, 246–249, 263, 265–268,
947–949, 951, 960–962, 1000, 1002, 271, 278, 296, 336, 338, 339, 343–345,
1015, 1025, 1040, 1067, 1103, 350, 358, 362, 365, 367, 369–371, 384,
1105–1107, 1118–1122, 1125–1130, 429, 431–434, 443, 444, 465, 466, 470,
1136, 1138–1151, 1158–1162, 1164, 472, 484, 487–489, 494–498, 501, 502,
1184, 1206, 1209–1212, 1237, 1241, 566, 567, 569, 570, 572, 574, 576–579,
1255, 1266–1269, 1271, 1272, 1274, 603, 605, 637, 640, 650, 654–656, 660,
1276–1278, 1280, 1281, 1305–1317, 665, 700, 701, 703, 704, 791, 808, 815,
1319, 1329–1337, 1339, 1340, 1357, 843, 863, 864, 871, 877, 894, 906, 917,
1369, 1393–1398, 1412, 1413, 1416, 918, 932, 939, 982, 986–989, 1036,
1458, 1459, 1470, 1473, 1480, 1047, 1067, 1081–1085, 1089, 1103,
1482–1485, 1487, 1496, 1497, 1117–1122, 1125, 1126, 1128–1130,
1500–1502, 1504, 1506, 1517–1519, 1142, 1145, 1146, 1148, 1150, 1151,
1521–1523, 1525–1528 1161, 1200, 1211, 1251, 1255, 1258,
Extraction methods, 591, 994, 995, 998–1002, 1267, 1274–1277, 1305, 1306,
1267, 1354 1310–1313, 1315–1317, 1329, 1330,
1334, 1348, 1349, 1351, 1354, 1369,
1393, 1395, 1397, 1405, 1407, 1414,
F 1417, 1423, 1430, 1443, 1459, 1464,
Ficus, 58, 72, 97, 99, 100, 110, 114, 137, 204, 1469, 1470, 1472, 1502
232, 236, 487–502, 839, 1040, 1238, FTIR analysis, 592, 593, 599, 709, 713,
1396, 1412 725, 943
Floral diversity, 330
Folk medicine, 107, 123–156, 216, 253, 548,
556, 1118, 1336, 1479 G
Food, 2, 10, 11, 21, 40, 44, 45, 107, 110, 123, Garlic, 12, 144, 153, 245–248, 265, 484, 1058,
127, 131, 132, 144, 149, 152, 159, 175, 1140, 1149, 1252, 1309, 1352, 1357,
183, 200, 210, 227, 234, 235, 237, 1358, 1480, 1482–1484, 1496–1497
243–251, 259, 271, 274, 280, 294, 296, Gas chromatography-mass spectrometry
322, 323, 330, 357, 359, 360, 370, 373, (GC-MS), 389, 390, 416–418, 446,
374, 401, 411, 413, 414, 418, 429, 450, 465, 650, 651, 654–656, 659, 733–735,
464, 466, 472, 473, 486, 488, 498, 502, 900, 943
Index 1539
N OD values, 600
Nagaland, 2, 3, 243, 250, 251 Ophiorrhiza brunonis, 727–736
Nano medicinal compounds, 1408, 1413, 1414 Orthosiphon, 81, 409–421
Nanoparticles, 448, 470, 554, 571, 573, 792,
810, 816, 1147, 1178–1189,
1271–1277, 1279–1281, 1366–1369, P
1371–1373, 1375, 1377, 1388–1401, Paliyar tribes, 44–47
1405–1423 Panax ginseng, 278, 777, 1084, 1252
Nanocarrier, 810, 813, 1180, 1278, 1368, Percolation, 995
1374, 1377–1379, 1409, 1410, 1415 Pharmacological, 102, 103, 240, 254, 256,
Nanofibre, 1369, 1376, 1377, 1379 259, 270, 272, 275, 278, 333, 351, 353,
Nanotechnology, 470, 472, 815, 816, 1177, 360–365, 367, 369–372, 381, 382, 395,
1187, 1190, 1194, 1242, 1277, 400, 403, 411, 414, 416, 421, 481–502,
1366–1368, 1371, 1387, 1408 513–514, 518, 522, 532–535, 557, 567,
Nardostachys jatamansi, 188, 311, 611–620, 590, 617, 624, 637, 649–686, 707, 708,
842, 937, 1038, 1238 727, 745, 746, 749, 754–756, 768, 778,
Natural antimicrobials, 605, 815, 1271, 1276 779, 784, 790, 794, 799, 823, 848, 857,
Natural compounds, 257, 318, 410, 420, 483, 880, 945, 961, 1037–1043, 1049, 1051,
605, 606, 757, 767, 783, 794, 798, 989, 1056, 1061–1072, 1097–1107, 1118,
1052, 1097, 1098, 1107, 1138, 1139, 1120, 1162, 1207, 1240, 1251, 1268,
1162, 1191, 1212, 1254, 1255, 1260, 1329, 1331, 1333, 1339, 1405, 1420,
1269, 1272, 1328, 1408, 1485, 1495, 1422, 1423, 1465–1469, 1475, 1488,
1496, 1499, 1518 1503, 1519, 1523
Natural drugs, 258, 309, 740, 743, 830, Pharmacological activities, 269, 272, 363, 365,
1061–1062 372, 374, 403, 411, 413, 414, 418, 419,
Natural products, 253, 254, 261, 308, 329, 429, 494, 501, 502, 511, 512, 518–522,
372, 409, 420, 481–484, 494, 679, 743, 529, 530, 538, 566, 573, 603, 612, 634,
745, 757, 767, 771, 777, 783, 784, 735, 745, 794–799, 942, 966, 1065,
789–790, 798, 808, 811, 815, 961, 994, 1068, 1125–1130, 1212, 1255, 1266,
1051, 1061, 1070, 1097, 1118, 1136, 1319, 1357, 1379, 1465
1162, 1207, 1250, 1254–1260, Pharmacological features, 1464
1265–1282, 1340, 1351, 1360, 1481, Pharmacology, 360–372, 381–383, 395–403,
1485, 1495, 1496, 1498, 1506, 466, 511–523, 768, 770, 853, 880, 929,
1507, 1523 989, 1067–1068
Necrosis, 400, 554, 570–573, 576, 615, 684, Phenolics, 269, 271, 273, 368, 369, 390, 391,
797, 978, 995, 1011, 1098, 1193, 1206, 395, 413, 414, 416, 465, 471, 484, 490,
1209, 1220, 1255, 1256, 1258, 1259, 501, 502, 533, 547, 550, 567, 569, 571,
1301, 1305, 1338, 1372, 1463, 1497, 589–591, 598, 601–604, 606, 623, 624,
1518, 1525–1527 634–643, 651, 655, 657, 662, 668, 670,
Nerve regeneration, 1331 712, 718, 719, 722, 731–732, 749, 753,
Neurodegenerative diseases, 412, 419, 483, 771, 774, 777, 778, 790, 793, 808–812,
631, 684, 798, 1202, 1236, 1237, 1327, 814–817, 864, 865, 869–872, 878, 910,
1328, 1335, 1337, 1340 929, 938–942, 948, 956, 960, 961, 966,
Nigella sativa, 1147, 1251, 1313, 1496 973, 974, 978, 980, 983, 987, 1049,
North Maharashtra, 25–40 1050, 1052, 1055, 1056, 1065–1068,
NO-synthase, 877, 1209 1070, 1072, 1101, 1102, 1105, 1107,
Nutraceuticals, 463–473, 546, 553, 704, 1120–1122, 1126, 1200, 1206, 1207,
739–757, 880, 973, 974, 979, 980, 989, 1209, 1210, 1218, 1219, 1255, 1258,
1055, 1118, 1251 1267, 1270, 1279, 1306, 1310, 1311,
1314, 1317, 1336, 1339, 1347,
1349–1352, 1449, 1466, 1496,
O 1498, 1502
Ocimum sanctum, 21, 110, 263, 266, 1147, Phytochemical analysis, 240, 366, 429, 708,
1280, 1304, 1319, 1516–1519, 1524, 709, 718, 719, 729–731, 733, 735, 929,
1526, 1528 938, 939, 967, 1121, 1418
1542 Index
Phytochemical constituents, 369, 382, 390, 624–629, 634, 636–638, 643, 649–651,
409, 410, 413–421, 547–548, 554, 557, 658, 663, 699–701, 703–705, 707–710,
703, 719, 966, 1049, 1051 712–716, 719, 722–725, 727–729,
Phytochemicals, 102, 103, 253, 272, 280, 292, 733–736, 740, 745, 767–785, 790, 791,
296, 348, 350–353, 360–365, 367, 369, 796, 807–812, 815–817, 830, 832, 833,
370, 372–374, 381–383, 385, 386, 836, 837, 840, 841, 843, 846, 847, 858,
389–391, 402, 403, 409, 411, 413, 414, 863–865, 869, 893–900, 905–924, 927,
416–419, 429–431, 448, 464, 465, 929–941, 943, 945–947, 951, 954, 961,
481–502, 545, 547, 549, 554, 565, 567, 962, 965–967, 971–989, 1002,
569, 571, 572, 574, 592, 602, 606, 640, 1005–1007, 1010–1012, 1022,
649–662, 703–704, 712–713, 718–723, 1026–1029, 1035–1043, 1047–1058,
725, 727, 728, 736, 774, 784, 815, 817, 1061–1068, 1070–1072, 1081–1088,
864, 871, 929, 930, 938, 939, 943, 947, 1102, 1103, 1105–1107, 1117–1123,
959, 961, 963, 966, 967, 978, 1125–1127, 1129, 1130, 1136,
1047–1049, 1056, 1057, 1061, 1072, 1138–1151, 1159–1163, 1191–1194,
1097, 1102, 1105, 1118–1125, 1129, 1199, 1200, 1206–1207, 1212, 1213,
1130, 1136, 1137, 1152–1158, 1216–1218, 1235, 1238–1241, 1248,
1160–1162, 1164, 1199, 1206, 1208, 1250, 1251, 1257, 1260, 1265–1271,
1212–1217, 1236, 1237, 1239, 1240, 1276, 1277, 1280, 1281, 1299–1319,
1242, 1281, 1318, 1328, 1329, 1335, 1328, 1329, 1335–1337, 1340, 1341,
1336, 1341, 1349, 1351, 1352, 1359, 1347–1360, 1366, 1368, 1369, 1374,
1360, 1365–1379, 1405, 1407–1410, 1376, 1379, 1387–1401, 1405–1423,
1414, 1415, 1421, 1422, 1448, 1449, 1448, 1449, 1457, 1461–1475,
1461, 1485, 1495–1497, 1517, 1520 1479–1481, 1484–1486, 1489, 1498,
Phytochemicals nanoencapsulated, 1272 1502–1506, 1516, 1518, 1519, 1523,
Phytochemistry, 360–372, 381–383, 386–395, 1524, 1526, 1528
429–430, 464–466, 483, 490–494, Pneumonia, 4, 132, 175, 194, 196, 228–233,
511–523, 530–532, 649–686, 929, 239, 240, 557, 606, 662, 678, 679,
938–939, 1120 1118, 1203, 1460, 1525
Phyto-constituents, 270, 272, 275, 278, 279, Poisonous medicinal plants, 1457–1475
421, 447, 547, 548, 572, 597, 602, 603, Polymeric nanoparticle (PNP), 1179–1180,
703, 712, 727, 729, 731–735, 821–858, 1184, 1374–1376, 1378, 1379,
938, 961, 1047, 1123–1125, 1130, 1398–1400
1200, 1206, 1251, 1253, 1257, 1473, Polyphenolics, 428, 496, 502, 591, 598,
1475, 1518, 1523 601–603, 605, 607, 650, 749–753, 811,
Phytomedicine, 463–473, 545–557, 932, 812, 815–816, 865, 871, 974, 1047,
1200, 1238 1056, 1101, 1122, 1130, 1209, 1217,
Phyto nanotechnology, 1408, 1410 1237, 1268, 1272, 1314, 1371,
Plant-based anticancer drug, 565, 1374 1423, 1449
Plants, 1–18, 20–22, 25, 27, 40, 43, 44, 50, 51, Polyphenols, 253, 269–271, 463–465, 467,
57, 58, 90–94, 96, 97, 99, 102, 103, 470, 487, 496, 547, 553, 603, 636, 640,
107–120, 123–153, 155, 159, 161, 744, 745, 749, 757, 780, 790, 807–817,
163–165, 172–217, 222, 223, 227, 228, 863–880, 938, 942, 943, 977, 978, 983,
230–235, 237–240, 244, 246–249, 253, 984, 987, 988, 1067, 1101, 1105, 1119,
254, 257–259, 265, 268–275, 278–280, 1138, 1139, 1206, 1216–1221, 1236,
287–289, 292, 298, 300, 308–318, 323, 1270, 1276, 1279, 1313, 1349,
329, 330, 333–335, 337, 339–346, 1422–1423, 1504, 1505
348–353, 357, 359–374, 381–383, Potential cyanide, 628, 630, 632, 633,
385–387, 395, 401–404, 409–414, 635, 642
416–421, 427–450, 463, 464, 466, 467, Preservation, 50, 259, 313, 315, 324, 330, 333,
469, 472, 473, 481–489, 495–497, 358, 411, 591, 604, 605, 753, 781,
500–502, 511, 513, 515, 516, 518, 519, 815–816, 988, 1102, 1348, 1486
529, 532, 545–549, 551–557, 566–575, Psidium guajava, 117, 235, 239, 1216,
589–591, 601, 602, 604–606, 612, 1251, 1420
Index 1543
Terminalia, 88, 94–97, 103, 151, 261, 263, 266, Traditional healers, 3, 43–103, 108, 109, 120,
271–272, 278, 313, 775, 779, 1008, 229, 290, 318, 320, 334, 1475, 1480
1011, 1015, 1023, 1026, 1117–1130, Traditional medicinal knowledge, 292
1211, 1277, 1280, 1306, 1317 Traditional medicinal plants, 229, 287, 352,
Therapeutic agent, 409, 419, 420, 448, 483, 353, 1486
496, 799, 1237, 1248, 1457 Traditional medicine, 1, 43, 44, 102, 156, 227,
Therapeutic efficacy, 280, 411, 781, 244, 253, 254, 256–258, 260, 264, 269,
1340, 1399 271, 272, 274, 275, 280, 281, 287, 288,
Therapeutic properties, 20, 253, 254, 264, 270, 307, 308, 310, 329, 330, 333, 386, 390,
272, 273, 386, 411, 482, 485, 782, 896, 409, 411, 414–416, 444, 481–484, 486,
937, 1049, 1238 502, 511, 522, 529, 538, 545–547, 557,
Therapeutics, 1–4, 8, 11, 16, 18, 20, 21, 253, 569, 573, 649, 735, 736, 739, 740, 767,
255–258, 261, 264, 270–272, 279, 309, 830, 858, 932, 935, 1035, 1036, 1041,
314, 318, 333, 382, 410, 413, 418–421, 1042, 1102, 1117, 1118, 1299, 1335,
430, 447, 448, 468, 471, 481, 483, 486, 1360, 1420, 1457, 1479, 1480, 1482,
487, 498, 501, 502, 519, 522, 523, 540, 1485, 1489, 1500
545, 553, 604, 616, 649, 684, 705, 727, Transcription factors (TFs), 390, 442, 447, 472,
735, 739, 744–745, 748, 768, 770–771, 614, 732, 734, 747, 795, 798, 1048,
779–782, 784, 789–800, 853, 858, 877, 1107, 1137, 1139, 1160, 1216, 1258
932, 933, 938, 939, 942, 975, 976, 980, Transgenesis, 1053
1029, 1040, 1043, 1047, 1049, 1051, Trans-Himalaya, 308, 310, 311, 314, 315,
1068, 1103, 1106, 1118, 1120–1122, 324, 1037
1125, 1127, 1130, 1137, 1161–1163, Transmembrane protease serine 2
1177, 1178, 1186, 1187, 1189, 1190, (TMPRSS2), 1520, 1528
1205, 1211, 1212, 1216, 1236, 1238, Tribes, 25, 44, 45, 98, 107–110, 119, 120, 227,
1239, 1248, 1250, 1251, 1256, 1258, 229, 243, 250, 386, 488, 701, 703, 710
1260, 1266, 1300, 1304, 1329, 1337, Trigonella foenum-graecum L., 38, 262, 268,
1341, 1358, 1375, 1389, 1391, 1401, 1252, 1358, 1496
1407, 1408, 1413, 1418, 1439, 1469, Trillium govanianum, 210, 381–404
1479, 1481, 1486, 1487, 1495–1507, Triphala, 263, 264, 1118, 1127, 1276
1515, 1521, 1522, 1528 Tumour cells, 536, 572, 574, 975, 1177, 1187,
Thymol, 363, 415, 660, 813, 814, 894, 1190, 1194, 1376, 1390–1392, 1400,
996–1002, 1050, 1066, 1267, 1269, 1407, 1438
1277, 1279, 1316, 1357, 1359
Tinosopra cordifolia, 272, 1518, 1519, 1523,
1524, 1526 U
Toxic plants, 939, 1475 Unani medicine, 255, 259, 272, 274, 294,
Traditional, 1–3, 21, 25, 26, 40, 44, 96, 97, 295, 1469
107, 108, 120, 123, 155, 163, Urtica dioica, 205, 1106–1107, 1149, 1273
227–230, 234–239, 243, 244,
249–251, 253–257, 259–261, 264,
269, 280, 281, 287–290, 296, 307, W
308, 310–313, 315–322, 324, 329, Wet tropics, 357–374
330, 333, 350, 352, 358–360, 374, Withania somnifera, 262, 263, 266, 272–273,
381, 382, 386–388, 410–414, 416, 345, 779, 847, 1150, 1200, 1238, 1255,
418, 421, 427, 429, 444, 446, 450, 1329–1331, 1516, 1518, 1520, 1524,
468, 485, 486, 488, 489, 496, 502, 1526, 1528
516, 522, 546, 552, 556, 633, 699,
707, 727, 739, 741, 743, 768, 770,
790, 830, 896, 929, 932, 933, 935, Z
936, 944, 945, 965, 979, 981, 1035, Zingiber officinale, 95, 96, 118, 153, 233, 238,
1036, 1043, 1049, 1054, 1057, 1061, 239, 263, 278, 428, 439, 448, 779,
1067, 1081, 1090, 1100, 1103, 1107, 1100, 1101, 1219, 1313, 1523
1118, 1128, 1162, 1180, 1212, 1259, Zingiberaceae, 4, 8, 11, 20, 69, 73, 111, 113,
1276, 1299, 1300, 1338, 1339, 1365, 118, 134, 135, 138, 141, 153, 208,
1367, 1388, 1395, 1473, 1479–1481, 230–233, 248, 427–450, 1140, 1145,
1520, 1522, 1524 1151, 1208, 1312, 1313, 1317