The Potential of Plant Extracts in Cell Therapy
The Potential of Plant Extracts in Cell Therapy
Abstract
Cell therapy is the frontier technology of biotechnology innovation and the most promising method for the treat-
ment of refractory diseases such as tumours. However, cell therapy has disadvantages, such as toxicity and poor
therapeutic effects. Plant extracts are natural, widely available, and contain active small molecule ingredients that are
widely used in the treatment of various diseases. By studying the effect of plant extracts on cell therapy, active plant
extracts that have positive significance in cell therapy can be discovered, and certain contributions to solving the cur-
rent problems of attenuation and adjuvant therapy in cell therapy can be made. Therefore, this article reviews the cur-
rently reported effects of plant extracts in stem cell therapy and immune cell therapy, especially the effects of plant
extracts on the proliferation and differentiation of mesenchymal stem cells and nerve stem cells and the potential
role of plant extracts in chimeric antigen receptor T-cell immunotherapy (CAR-T) and T-cell receptor modified T-cell
immunotherapy (TCR-T), in the hope of encouraging further research and clinical application of plant extracts in cell
therapy.
Keywords: Cell therapy, Plant extracts, Stem cell, CAR-T, TCR-T
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativeco
mmons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 2 of 14
and isoproterenol), dietary supplements/medicinal The effect of plant extracts on stem cell therapy
plant extracts (e.g. berberine), and endogenous drugs The effect of plant extracts on MSCs
(e.g. estrogen) [8, 9]. Among them, plant extracts are an MSCs exist in almost all postnatal human tissues. The
important source of bioactive small molecules, which major sources of adult MSCs are mainly from bone mar-
are mainly derived from herbal plants, and include flavo- row, adipose tissue, etc. [7, 15]. Compared with ESCs,
noids, alkaloids, polysaccharides, volatile oils, etc. Plant MSCs are easier to isolate and culture in vitro, and more
extracts play an important role in disease treatment, importantly, there are fewer ethical issues. Furthermore,
especially in cancer and infectious diseases. Herbal ther- due to their HLA-DR-negative feature, MSCs do not have
apy is a traditional medical practice that has long been immunogenic in therapy [7]. MSCs are characterized
used to treat a variety of diseases. Herbal medicine is safe by different sources, isolation methods, and epigenetic
and affordable [7]. It is a promising alternative approach changes during growth. They can be differentiated into
with a significant effect on alleviating patient disease. osteocytes, neurons, and angiogenesis, through stimula-
Therefore, studying the effects of plant extracts in cell tion with plant extracts (Fig. 1).
therapy and digging out active plant extracts in herbal
medicines can provide guidance for adjuvant therapy Proliferation effect
combined with cell therapy. Many plant extracts, such as Foeniculum vulgare [16],
The application of plant extracts in cell therapy has Ferula gummosa [17], amentoflavone (Selaginella tam-
gradually increased with the rapid development of cell ariscina (P. Beauv.) Spring) [18], gastrodin (Gastrodia
therapy. For example, the increased therapeutic effects of elata Bl.) [19], and resveratrol (Polygonum cuspida-
stem cells induced by plant extracts have been reported tum Sieb. et Zucc.) [20], can significantly increase bone
in studies of Alzheimer’s disease [10], chronic kidney marrow-derived human MSCs (BM-hMSCs) prolifera-
disease [11], and stroke [12], and the therapeutic effects tion. In addition, ginsenoside Rg1, which is an effective
of immune cells induced by plant extracts have been compound in Panax ginseng, Panax notoginseng, and
reported in diseases such as non-small-cell lung cancer American ginseng, can also promote cell proliferation
and liver cancer [13, 14]. This review describes the effects [21]. Apple ethanol extract promotes proliferation of
of plant extracts, mainly herbal extracts, in cell therapy hASCs and human cord blood-derived MSCs via ERK
approaches and clarifies the potential mechanisms of signalling [22]. Tinospora cordifolia and Withania som-
action when possible. nifera are traditional Ayurveda medicinal materials in
Fig. 1 Sources of MSCs and their proliferation, differentiation, angiogenesis, antilipogenesis, and antioxidant stress effects stimulated by plant
extracts
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 3 of 14
India that are reported to improve cell proliferation Osteogenic effects through Wnt signalling pathways
ability and activity, as well as reduce cell apoptosis and Flavonoids of epimedii(Epimedium brevicornum Maxim.,
postpone aging [23]. ZD-I is a prescription composed etc.) were found to increase the rates of osteogenic activ-
of seven traditional Chinese medicines (TCM). It has ity through the BMP or Wnt-signalling pathway [38]. In
stimulatory effects on the proliferation of hMSCs [24]. addition, Angelica sinensis polysaccharide can enhance
Viscum album induces primitive placenta-derived the osteogenic differentiation of rat BM-MSCs cultured
MSCs (PDSCs) with remarkable proliferative properties in high-sugar and guide bone regeneration in type 2 dia-
through autophagy mechanism. Specifically, Viscum betes animal model which chained to the Wnt/β-catenin
album can regulate the cell cycle to make PDSCs self- signalling pathway [39]. Ginkgo biloba and its main com-
renewal and regulate the induction of survival factors, ponent ginkgolide B accelerate osteoblast differentiation
apoptosis and autophagy to reduce cell death [25]. and the formation of bone via Wnt/β-Catenin signalling
[40] (Fig. 2). Berberine (Coptis chinensis Franch.) [41] and
salvianolic acid B (Salvia miltiorrhiza Bge.) [42, 43] pro-
Differentiation effect mote osteogenesis in BM-MSCs through Wnt/β-catenin
Osteogenic effects signalling and strengthen Runx2 expression. Salvianolic
Osteogenic effects via transcription factors acid B influences the ERK signalling pathway and lower
Foeniculum vulgare, Ferula gummosa, and amentofla- PPARγ mRNA, accelerating the osteogenesis of MSCs.
vone can significantly increase the alkaline phosphatase The osteogenic-related genes can be strengthened under
(ALP) activity of hMSCs and promote BM-hMSC dif- the induction of naringin, and the expression of Notch1
ferentiation into osteoblasts [16–18]. Dipsacus asper can be up-regulated at the same time, and activation Wnt
and its ingredients hedraganin-3-O-(2-O-acetyl)-α-l- signalling activation [44].
arabinopyranoside enhance osteoblastic differentiation
not only by inducing ALP activity but also by induc-
ing bone sialoprotein and osteocalcin expression [26]. Osteogenic effects through BMP signalling pathways
Moreover, Fructus Ligustri Lucidi effectively activated Ginkgo biloba has been found to enhanced Runx2
ALP, reduced the osteogenic differentiation time of expression and regulated BMP4 in BMP signalling [45].
MSCs, and up-regulated the expression of osteogenic Moreover, harmine [33], silibinin(Silybum marianum
related factors such as catenin, BMP2, cyclin D1, mem- (L.) Gaertn.) [46], and genistein(Genista tinctoria Linn.,
brane matrix metalloproteinase, osteoprotegerin and etc.) [47] activate the BMP and Runx2 pathways. Duhuo
T-box 3 [27]. Poncirin (Poncirus trifoliata (L.) Raf ) Jisheng decoction and its effective component Ligusticum
[28], Panax notoginseng saponins [29, 30], and naringin Chuanxiong can activate Smad 1/5/8 and ERK signal-
(Citrus grandis) [31, 32] decreased peroxisome prolif- ling, increase the osteogenic effect of MSCs, and improve
erator-activated receptor γ (PPARγ) 2 mRNA levels, BMP-2 and Runx2 [48] (Fig. 2).
while Panax notoginseng saponins raised the levels of
ALP, Cbfa 1, OC, BSP, OPG, β-catenin, and cyclin D1.
Harmine (Peganum harmala L.) increased ALP activ- Osteogenic effects through MAPK signalling pathways
ity and up-regulated osteocalcin expression. Moreover, Most of the plant extracts used in the study of MSCs
harmine can up-regulate osterix [33]. The combination osteogenesis are TCM monomers. Icariin (Epimedium
of epigallocatechin-3-gallate [34] and bone inducer brevicornum Maxim., etc.), amentoflavone and quercetin
can up-regulate BMP2 and enhance bone formation. promoted osteogenesis via the JNK and p38 MAPK path-
Gastrodin [19] improved ALP, OCN, COL I, and OPN ways (Fig. 2). Icariin also phosphorylates ERK, and stimu-
while reducing ROS. Fucoidan enhanced osteogenic lates PI3K-AKT-eNOS-NO-cGMP-PKG pathway in bone
specific marker genes, such as ALP, osteopontin, type I marrow stromal cells [18, 49, 50]. Quercetin is a flavonoid
collagen, Runx2, and osteocalcin in ASCs [35]. Querce- that can also activate ERK signalling pathways, decrease
tin (Sophora flavescens Ait.) can increase Osx, Runx2, the aging and oxidative stress in MSCs, and promote
BMP2, Col1, OPN and OCN, and enhance osteo- osteogenic differentiation [51, 52]. Fucoidan can induce
genic differentiation [36]. BuShenNingXin decoction osteogenic differentiation, activate ERK and JNK mainly
(BSNXD) up-regulated ALP and collagen type I, osteo- through BMP2 Smad 1/5/8 signalling, and regulate osteo-
calcin, Runx2, and osterix. Furthermore, BSNXD was genic differentiation markers [53, 54]. One study showed
shown to reduce the quantity of adipocyte and PPARγ that resveratrol enhanced cell renewing by inhibiting cell
mRNA [37]. A summary table of plant extracts that aging at a low concentration, while it inhibits cell self-
stimulate osteogenesis of MSCs is shown in Additional renewal by up-regulating cell senescence, doubling time,
file 1: Table S1. and S-phase arrest at a high concentration. In addition, it
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 4 of 14
Fig. 2 Plant extracts that affect MSCs osteogenesis by regulating intracellular signalling pathways. Ginkgolide B, Panax notoginseng saponins,
berberine, and salvianolic acid B regulate axin, β-catenin, and TCF in the Wnt signalling pathway; Ginkgo biloba, harmine, silibinin, genistein, and
Ligusticum chuanxiong regulate BMP, Runx2 and Smad 1/5/8 in the BMP signalling pathway; Resveratrol, icariin, amentoflavone, quercetin, and
fucoidan regulate p38, ERK1/2, and JNK in the MAPK signalling pathway
can stimulate MSCs and promote osteoblast differentia- upregulate NSE, MAP-2, GAP-43, NCAM, and SYN-1
tion by acting on ER-dependent mechanisms and activat- genes [21]. Another study showed that gisenoside Rg1
ing ERK1/2 [20, 55, 56]. can promote neural differentiation in mouse ASCs by
miRNA-124 signalling pathway [63]. Similarly, radix
Neurogenic effects Angelicae sinensis can also induce adipose-derived MSCs
Mucuna gigantea grows natively in Hawai ‘i. It was to differentiate into neuron-like cells [64]. In Ayurvedic
recorded that it can be used to treat kampavata (excita- medicine, dhanwantharam kashayam is considered a
tory paralysis) [57]. Mucuna gigantea can promote growth stimulant for children, which can promote nerve
proliferation feature, nestin, and β-III tubulin mRNA regeneration [65].
expression in MSCs [58].
A study using human umbilical cord Wharton’s Jelly- Angiogenesis effects
derived MSCs (WS-MSCs) showed that Salvia miltio- Treatment of hMSCs with olive leaf extract promoted the
rrhiza increases the expression of nestin, β-tubulin, differentiation of cells into endothelial cells and develop-
neurofilament, GFAP, and neurite outgrowth-promoting ment of the tubular construction needed for angiogen-
protein [59]. Another study in rat BM-MSCs showed esis. At the same time, olive leaf extract can promote
that Salvia miltiorrhiza promotes Mash-1 and NGN-1 VEGF, PCAM, PDGF, and VEGFR-1 [7]. Curcumin is an
induced mRNA expression of TUJ-1, NF, and synapto- antioxidant and anti-inflammatory substance in turmeric.
physin [60]. Its ethanol extracts can increase the expression of CD34,
Ginkgolide B and Astragalus mongholicus can increase CD133, and VEGFR2 to cause ASCs to proliferation and
NSE-positive neuron-like cells and GFAP-positive astro- differentiation into endothelial progenitor cells [66]. In
cyte-like cells to promote MSCs differentiation into the animal hindlimb ischaemia model, fucoidan can pro-
nerve cells [61, 62]. Moreover, Astragalus mongholi- tect MSCs from oxidative stress and enhance angiogen-
cus also enhances the expression of the Wnt-1 gene and esis. Another study showed that fucoidan can inhibit the
Ngn-1 gene [62]. Ginsenoside Rg1 was found to acceler- cell death caused by MSCs ischemia, and adjust the lev-
ate the differentiation of neural phenotype in hASCs and els of apoptosis-related proteins and cellular ROS mainly
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 5 of 14
by MnSOD and Akt pathways [67]. In addition, through MSCs. However, some studies have demonstrated that
ERK-IDO-1 signalling cascade, it increases the prolifera- plant extracts have side effects. Cimicifugae Rhizoma,
tion potential and the expression of cell cycle-associated also called Shengma in China, affects the vita of dental
proteins, and enhances the immunoregulation activity of stem cells, and has side effects on the oral cavity at a high
MSCs [11]. Carica papaya leaf extract, rich in papain, was content [78]. Additionally, Asiasarum radix is the same as
found to enhance the composition of IL-6 and stem cell Cimicifugae Rhizoma [79]. As mentioned above, Fructus
factors related to platelet production in vitro [68]. Ligustri Lucidi has an osteogenesis effect. Nevertheless,
Fructus Ligustri Lucidi in a dose of more than 200 μg/mL
Anti‑adipogenic effects has cytotoxicity to MSCs [27].
Some plant extracts also have anti-adipogenic effects
on MSCs. The results of one study confirmed that the
The effect of plant extracts on NSCs
antioxidant action of Tithonia diversifolia may influ-
Endogenous NSCs are abundantly in the subventricular
ence the expression of HO-1. More importantly, it may
region of the hippocampal granular area and the germinal
regulate carbohydrate and fat metabolism by repressing
area of the cerebrum. They differentiate cells according to
adipocyte differentiation through activating AMPK [69].
the needs of brain structure and function [80]. NSCs are
In an experiment using MSCs, after stimulation with
primarily used to remedy central nervous system injury
aloe-emodin, many indicators were reduced, including
and degenerative diseases. There are two intervention
resistin, adiponectin, aP(2), lipoprotein lipase, PPARγ,
strategies involving NSCs. One strategy involves using
and TNFα, which influence adipogenic pathways [70].
endogenous NSCs to repair the diseased site, but endog-
Quzhisu can repress adipogenic differentiation of BM-
enous NSCs are not sufficient and prefer to differentiate
MSCs by downregulating PPARγ [71]. Similarly, flavo-
into gliocytes instead of neurons; the other strategy is
noids of epimedii like Quzhisu downregulate PPARγ, and
transplantation of exogenous NSCs [61]. However, it is
can also decrease C/EBP-α [72].
difficult to control the survival, replication, and differen-
tiation of exogenous NSCs into local nerve cells. Sources
Antioxidant stress effects
of NSCs include human ESCs, human iPSCs, human foe-
Undaria pinnatifida, Myrtus community L. and Cirsium
tal brain-derived neural stem/progenitor cells, and direct
setidens showed antioxidant stress effects in MSCs. Und-
reprogramming of astrocytes. According to their func-
aria pinnatifida, also called Mi-Yoek in Korea, is con-
tions, they can be divided into pluripotent and multi-
sidered a healthy food. The anti-aging effect of Undaria
potent cell types [81]. Plant extracts play an important
pinnatifida in BM-MSCs was researched. The results
part in promoting the proliferation and differentiation
showed that after H2O2 treatment, it had the effect of
of NSCs into new neurons. The Notch, Wnt, BMP, and
antioxidant stress, and could decrease aging and improve
sonic hedgehog signalling pathways have been the most
the differentiation potential of cells by controlling ROS
studied [82].
[73]. In addition, icariin protected rabbit BM-MSCs from
oxygen, glucose, and apoptosis via inhibition of ERs-
mediated autophagy associated with MAPK signalling Proliferation effect
[74]. Furthermore, residues from the production of Myr- Ginsenosides Rg1 advances the incorporation of Bromo-
tus community L. can counteract the appearance of aging 2-deoxyuridine and the expression of nestin and vimen-
phenotypes in ASCs, reduce oxidative stress and inflam- tin in NSCs, and promotes the proliferation of NSCs
mation, and enhance the expression of genes related to [83]. In addition, ginsenoside Rd can enhance the prolif-
pluripotency [75]. The authors also studied the genetic eration of NSCs in vivo and in vitro. It can enhance the
programs responsible for cellular senescence in human size and quantity of neurospheres [84]. After oxygen and
ASCs exposed to oxidative stress and found that in the glucose deprivation (OGD) /r injury in vitro, resveratrol
cells stimulated by Myrtle, the SA-β-Gal positive cells up-regulated the survival and proliferation of NSCs, and
and the cell cycle regulation genes were decreased, while increased patched-1, smoothened (SMO) and Gli-1 [85].
TERT and c-Myc genes were increased [76]. Cirsium Meanwhile, resveratrol can reduce the damage and raise
setidens [77] has a suppressive effect on cell injury by reg- the proliferation of NSCs by promoting Nrf2, HO-1 and
ulating oxidative stress and repressing apoptosis-related NQO1 [86]. Artesunate is a derivative of artemisinin
signalling pathways. from Artemisia annua [87]. It can inhibit transcription by
inducing Foxo-3a phosphorylation, then downregulating
Adverse p27kip1, and enhancing the proliferation of NSCs in the
The above studies have shown that certain plant extracts infarcted cortex through PI3K/AKT signalling transduc-
can promote the proliferation and differentiation of tion [88].
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 6 of 14
ESCs by promoting brain-derived neurotrophic factor/ damage to human keratinocyte stem cells, and effectively
tropomyosin receptor kinase B signalling pathway [109]. enriched the p53- specific ligasing ability of the mouse
double minute 2 homologue in UVB irradiation-induced
The effect of plant extracts on iPSCs p53 activation. Likewise, zingerone (Zingiber officinale
iPSCs have characteristics similar to those of ESCs in Rosc.) [125] can protect the epidermis by restraining the
terms of unlimited self-renewal and differentiation UV damage mediated by p42/44 MAPK and p38 MAPK.
capabilities. Plant extracts induce iPSCs production In other stem cells, Ginkgo biloba[126, 127] activates tel-
and apoptosis. The Sagunja-tang herbal formula can omerase through PI3k/Akt signalling pathway to delay
efficiently produce iPSCs from human foreskin fibro- the aging of endothelial progenitor cells. Additionally,
blasts via transcription factors [110]. Prunellae Spica starting from telomerase, TSY-1 [128] increases telomer-
and Magnoliae cortex-mediated apoptosis of undiffer- ase activity in CD34+ haematopoietic stem cells.
entiated iPSCs was found to be p53-dependent, and to
have potent anti-teratoma activity, with no genotoxicity Immune cell therapies
toward differentiated cells. Therefore, these compounds Adoptive cell therapy (ACT) is a kind of immunotherapy
can be used for iPSC-based cell therapy to induce apop- that is genetically modified T-cells to deliver a CAR or
tosis of possible undifferentiated iPSCs and prevent the TCR. To a certain extent, mutated cancer cells provide
occurrence of teratomas [111, 112]. many peptides that are not found in natural cells, which
Plant extracts can induce differentiation of iPSCs into brings a potential target for constructing a new antigen
nerve cells. Salvia miltiorrhiza can significantly increase screening system, and promotes the development of
the expression of nestin and microtubule-associated pro- ACT, making CAR-T and TCR-T treatment become the
tein 2 (MAP2) genes and proteins, and induce the dif- most prospective way to treat cancer. However, ACT has
ferentiation of iPSCs into neurons [113]. Plant extracts great differences in the treatment of various tumours
also have an improved effect on the nerve cell model dif- types, and there are still some shortcomings that need
ferentiated from iPSCs. N-Butylidenephthalide (n-BP) is improvement [129].
derived from Angelica Sinensis. N-BP can reduce Aβ40
deposits, total tau protein, and its hyperphosphorylated CAR‑T
form in iPSCs-derived neurons induced by Down syn- CAR is a kind of engineering, which can enable lympho-
drome [114]. Graptopetalum paraguayense can improve cytes to identify and eliminate cells delivering homolo-
AD-related phenotypes, such as reducing Aβ 40, Aβ 42, gous target ligands. It has antigen binding domain, hinge,
and tau protein phosphorylation [115]. transmembrane domain and intracellular signal domain
iPSCs are differentiated into cardiomyocytes, which modules. By changing each component, its function and
are used in the research of related diseases. One study anti-tumour effect can be adjusted. At present, vari-
found that Salvia miltiorrhiza and Crataegus pentagyna ous types of CARs are being developed and designed to
have anti-arrhythmic effects. Salvia miltiorrhiza has an improve the safety and effectiveness in cancer treat-
antioxidant effect, regulates calcium treatment on myo- ment [130]. Clinically, treatment with CAR-T-cells first
cardial cells during I/R and decreases arrhythmia and requires T-cells, which can be obtained from the patient’s
apoptosis [116]. Crataegus pentagyna extract has an peripheral blood, or allogeneic CAR-T-cells obtained
anti-arrhythmic effect on cardiomyocytes derived from from a donor [131]. T-cells are stimulated and expanded
human arrhythmia-specific iPSCs [117]. In addition, Yix- in vitro and transduced with specific CAR genes through
inshu capsule has a protective effect on human iPSCs- viral vectors, and then, the CAR-T-cells are infused back
derived cardiomyocytes by reducing endothelin 1 (ET-1) into the patient to perform the set tumour-killing effect
induced contractile dysfunction, increasing brain natriu- in the patient’s body. This type of therapy is also called
retic peptide (BNP) content, and inducing morphological CAR-T-cell therapy (Fig. 3).
changes [118]. However, some plant extracts are toxic to In 2017, the FDA approved two types anti-CD19 CAR-
cardiomyocytes, such as liensinine and neferine [119], T-cell products to treat both B-cell ALL and diffuse large
mitragynine [120], and Erythrina senegalensis DC [121]. B-cell lymphoma, and these products have transformed
the field of anticancer immunotherapy [134]. However,
The effect of plant extracts on other stem cells there are still some limitations in CAR-T-cell therapy.
Ultraviolet-B (UVB) irradiation can damage the epider- Mechanisms hampering CAR-T-cell efficiency include
mis. Andrographis paniculata [122] promotes the prolif- limited T-cell persistence and therapy-related toxicity.
eration of epidermal stem cells (EpSCs) and anti-aging via Furthermore, severe toxicities, restricted trafficking to,
increasing integrin β1 and VEGF expression. Morin [123] infiltration into and activation within tumours, antigen
and Vanillin [124] significantly inhibited UVB-induced escape and heterogeneity; manufacturing issues; physical
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 8 of 14
Fig. 3 CAR-T-cell therapy and the four generations of improvements. The first-generation CARs were fused with a single-chain variable fragment
(scFv) to a transmembrane domain and an intracellular signalling unit: the CD3 zeta chain. Then, the second-generation CARs improved the
costimulatory molecule receptor-like CD28, which is the most commonly used. The second-generation CARs increased the production of cytokines
and enhanced durability. The third-generation of CARs design in-corporated an additional costimulatory domain to enhance CAR function and
included the scFv, the initial CD3ζ-chain, and the CD28 and 4-1BB or OX40 costimulatory domains [132]. At present, fourth-generation CAR-T
therapy has been extended. In this type of CAR-T-cell therapy, cytokine genes have been added to the structure, which can stimulate high cytokines
expression that enhances the activity of T-cells after CAR-T-cells are activated, thereby improving the antitumour activity of CAR-T-cells [133]
properties; and the immunosuppressive capacities of [159], Lycium barbarum [160], Ganoderma lucidum
solid tumours have prevented the success of CAR-T- [161], Yunzhi-Danshen [162] can upregulate CD3+,
+ +
cells in these entities [135, 136]. Additionally, it may not CD4 , CD4 + /CD8 + and NK cells. Moreover, gastro-
be possible to obtain a sufficient number of T-cells from din was found to ameliorated the CD8+ T-cell-mediated
the patient because the patient is usually not considered immune response and significantly improved protection
for CAR-T-cell therapy, usually due to a reduction in the in tumour-challenged animals. This finding indicates that
number of original lymphocytes caused by previous cyto- gastrodin is a potential adjuvant contributing to antican-
toxic treatment [134]. In application, almost all CAR-T- cer immunomodulation.
cell products are derived from CD4+ T-cells and CD8+ On the other hand, the tumour microenvironment
T-cells, both cell populations likely contribute to treat- is a complex pathological system composed of tumour
ment effect [134]. Some plant extracts have beneficial cells, blood/lymphatic vessels, tumour stroma, and
effects on CD4+ T-cells and C D8+ T-cells. For example, tumour-infiltrating myeloid precursors, providing a liv-
Fuzheng Qingjie [137, 138], Fuzheng Fangai [139], Xiaoji ing environment for tumour cells and promoting tumour
[13], Cistanche deserticola [140], Epimedium koreanum metastasis. In the tumour microenvironment, tumour-
Nakai [141], Glycyrrhiza uralensis [142, 143], Aidi [144], infiltrating myeloid precursors mainly include tumour-
and Scolopendra subspinipes [145–147] can increase in associated macrophages, tumour-associated dendritic
CD4+ cells and the CD4/CD8 ratio, and produce FN-g, cells, and myeloid-derived suppressor cells, which inhibit
IL-2, IL-4, IL-6, and IL-7; Xiao Ai Ping [148], Lycium T-cells or other immune cells and play an important
barbarum [149–151], Dangguibuxue tang [152], Olden- role in its antitumour activity. Therefore, improving the
landia diffusa [153–155], Carthamus tinctorius [156], tumour microenvironment by targeting these cells is an
lectin-55 [157], and Tricosanthes kirilow [158] have an effective way to assist CAR-T-cell therapy. Liu J et al.
effect on increasing in CD8+ cells, and tumour infiltra- reviewed Chinese herbal medicine and its components
tion and increasing IFN-g and IL-10. Shenqi Fuzheng that induce tumour cell apoptosis and directly inhibit
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 9 of 14
tumour growth and invasion, providing new research present, cell therapy is promising. However, to under-
ideas for cell therapy [163]. stand the long-term effects, more in-depth research on
the dose and side effects of plant extract applications is
TCR‑T still needed. Although plant extracts are recognized as
Due to the limitations of CAR in the application, it only excellent alternatives to synthetic interventions, clini-
recognizes cell surface protein antigens, while TCR can cal application is challenging due to the variability and
distinguish intracellular proteins expressed as peptides complexity of the bioactive components present in the
on MHC class I molecules. Therefore, TCR-T therapy extracts, as well as the effects of solvents during extrac-
has superiority in the field of solid tumour treatment. tion. Therefore, the effects of plant extracts on cell ther-
The TCR can be produced in two ways. One method is to apy need to be better and more deeply researched to
identify and clone T- cells from patients with antitumour supplement the current deficiencies in cell therapy.
reactions. Their TCRs are inserted into retroviruses or
lentiviruses to infect target T-cells. Another method is Abbreviations
to isolate TCRs from humanized mice that recognize MSCs: Mesenchymal stem cells; NSCs: Nerve stem cells; CAR-T: Chimeric
tumour antigens. TCRs can be immunized with appro- antigen receptor T-cell immunotherapy; TCR-T: T-cell receptor modified T-cell
immunotherapy; ASCs: Adipose-derived stem cells; iPSCs: Induced pluripotent
priate tumour antigens because they can express human stem cells; ESCs: Embryonic stem cells; HLA-DR: Human leukocyte antigen-
MHC class I or II. After T-cells were isolated, the TCR antigen D related; BM-hMSCs: Bone marrow-derived human MSCs; ERK:
gene was cloned into a recombinant vector for genetic Extracellular signal-regulated kinase; Runx2: Runt-related transcription factor
2; HGPS: Hutchinson–Gilford progeria syndrome; VEGF: Vascular endothelial
engineering transformation of patients’ autologous growth factor; VEGFR: Vascular endothelium growth factor receptor; TCM:
T-cells [164]. Traditional Chinese medicine; ALP: Alkaline phosphatase; PPARγ: Peroxi-
Although effective responses have been observed in some proliferator activated receptor γ; BSNXD: BuShenNingXin decoction;
WS-MSCs: Wharton’s jelly-derived MSCs; C/EBP-α: CCAAT enhancer-binding
TCR-T-cell therapy, adverse reactions have become a protein-α; Smo: Smoothened; OGD: Oxygen and glucose deprivation; bFGF:
thorny issue in many trials. Most of the reasons are that Basic fibroblast growth factor; NSPC: Neural stem/progenitor cells; GFAP: Glial
TCR-T- cells, in addition to their killing effect on tumour fibrillary acidic protein; n-BP: Butylidenephthalide; ET-1: Endothelin 1; BNP:
Brain natriuretic peptide; UVB: Ultraviolet-B; EpSCs: Epidermal stem cells; ACT
cells, severely destroy normal cells with the same antigen : Adoptive cell therapy.
[129]. Since TCR-T-cells have only emerged in recent
years, there are almost no plant extracts currently used Supplementary Information
in TCR-T-cell research. Parvifoline AA is an ent-kaurane The online version contains supplementary material available at https://doi.
diterpenoid and can significantly stimulate the level of org/10.1186/s13287-022-03152-z.
NKG2D ligands on hepatocellular carcinoma cells, evi-
dently enhancing their recognition and lysis by NK cells Additional file 1: Table S1. Plant Extracts for the Osteogenesis of MSCs.
[14]. Perhaps improving the efficacy of TCR-T-cells in the
immunosuppressive microenvironment and determining Acknowledgements
that the expression is mainly (if not completely) limited We thank Lei Tong for guidance in drawing from University of science and
technology Beijing.
to cancer cell targets may be a future research direction
for plant extracts. Author contributions
H.J Yang, X.Y. Li and P. Chen designed the idea of this review; C.F. Li, Zh. Cui,
S.W. Deng and P. Chen co-wrote the paper with input from all authors. All
Conclusion authors read and approved the final manuscript.
Plant extracts are relatively easy to obtain and have
Funding
significant activity in the treatment of many diseases. This study was supported by fundamental Research Funds for the Central pub-
The above review shows that plant extracts have an lic welfare research institutes of China (ZZ13-YQ-082-C1; JBGS2021001), the
effect on stem cell proliferation or directed differenti- Scientific and Technological Innovation Project of China Academy of Chinese
Medical Sciences (CI2021A00610).
ation and play an important role in solving the prob-
lem of insufficient endogenous stem cells and directed Availability of data and materials
differentiation of stem cells; In immune cell therapy, Not applicable.
the effect of plant extracts on stem cells are reflected
in the beneficial effects on C D4+ T-cells and C D8+ Declarations
T-cells and the improvement of the tumour microen- Ethics approval and consent to participate
vironment. Moreover, plant extracts, such as astra- Not applicable.
galoside [165], paeoniflorin [166], and licorice [167],
Consent for publication
have a good immunoregulatory and anti-inflammatory Not applicable.
activities and may provide a better treatment plan for
the cytokine storm caused by cell therapy [168]. At
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 10 of 14
Competing interests 15. Brown C, McKee C, Bakshi S, Walker K, Hakman E, Halassy S, et al. Mesen-
All authors declare no competing interests. chymal stem cells: cell therapy and regeneration potential. J Tissue Eng
Regen Med. 2019;13:1738–55. https://doi.org/10.1002/term.2914.
Author details 16. Mahmoudi Z, Soleimani M, Saidi A, Khamisipour G, Azizsoltani A. Effects
1
Beijing Key Laboratory of Traditional Chinese Medicine Basic Research On of Foeniculum vulgare ethanol extract on osteogenesis in human
Prevention and Treatment for Major Diseases, Experimental Research Center, mecenchymal stem cells. Avicenna J Phytomed. 2013;3:135–42.
China Academy of Chinese Medical Sciences, Beijing 100700, China. 2 Institute 17. Mahmoudi Z, Soleimani M, Saidi A, Iranshahi M, Azizsoltanli A. Effect of
of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Ferula gummosa ethanolic extract on osteogenesis in human mesen-
Beijing 100700, China. 3 Robot Intelligent Laboratory of Traditional Chinese chymal stem cells. J Med Plants. 2013;12:Pe50–9.
Medicine, Experimental Research Center, China Academy of Chinese Medical 18. Zha X, Xu Z, Liu Y, Xu L, Huang H, Zhang J, et al. Amentoflavone
Sciences and MEGAROBO, Beijing, China. enhances osteogenesis of human mesenchymal stem cells through
JNK and p38 MAPK pathways. J Nat Med. 2016;70:634–44. https://doi.
Received: 6 September 2021 Accepted: 23 May 2022 org/10.1007/s11418-016-0993-1.
19. Huang Q, Shi J, Gao B, Zhang H-Y, Fan J, Li X-J, et al. Gastrodin: an
ancient Chinese herbal medicine as a source for anti-osteoporosis
agents via reducing reactive oxygen species. Bone. 2015;73:132–44.
https://doi.org/10.1016/j.bone.2014.12.059.
References 20. Dai Z, Li Y, Quarles LD, Song T, Pan W, Zhou H, et al. Resveratrol
1. Facklam AL, Volpatti LR, Anderson DG. Biomaterials for personalized cell enhances proliferation and osteoblastic differentiation in human mes-
therapy. Adv Mater. 2020;32: e1902005. https://doi.org/10.1002/adma. enchymal stem cells via ER-dependent ERK1/2 activation. Phytomedi-
201902005. cine. 2007;14:806–14. https://doi.org/10.1016/j.phymed.2007.04.003.
2. Bacakova L, Zarubova J, Travnickova M, Musilkova J, Pajorova J, Slepicka 21. Xu F-T, Li H-M, Yin Q-S, Cui S-E, Liu D-L, Nan H, et al. Effect of ginse-
P, et al. Stem cells: their source, potency and use in regenerative thera- noside Rg1 on proliferation and neural phenotype differentiation of
pies with focus on adipose-derived stem cells - a review. Biotechnol human adipose-derived stem cells in vitro. Can J Physiol Pharmacol.
Adv. 2018;36:1111–26. https://doi.org/10.1016/j.biotechadv.2018.03. 2014;92:467–75. https://doi.org/10.1139/cjpp-2013-0377.
011. 22. Lee J, Shin MS, Kim MO, Jang S, Oh SW, Kang M, et al. Apple ethanol
3. Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. extract promotes proliferation of human adult stem cells, which
Stem cell therapy in the heart: biomaterials as a key route. Tissue Cell. involves the regenerative potential of stem cells. Nutr Res. 2016;36:925–
2021;71: 101504. https://doi.org/10.1016/j.tice.2021.101504. 36. https://doi.org/10.1016/j.nutres.2016.06.010.
4. Buzhor E, Leshansky L, Blumenthal J, Barash H, Warshawsky D, Mazor Y, 23. Sanap A, Chandravanshi B, Shah T, Tillu G, Dhanushkodi A, Bhonde R,
et al. Cell-based therapy approaches: the hope for incurable diseases. et al. Herbal pre-conditioning induces proliferation and delays senes-
Regen Med. 2014;9:649–72. https://doi.org/10.2217/rme.14.35. cence in Wharton’s Jelly Mesenchymal stem cells. Biomed Pharmaco-
5. Goradel NH, Hour FG, Negahdari B, Malekshahi ZV, Hashemzehi M, ther. 2017;93:772–8. https://doi.org/10.1016/j.biopha.2017.06.107.
Masoudifar A, et al. Stem cell therapy: a new therapeutic option for 24. Chen M, Feng W, Cao H, Zou L, Chen C, Baatrup A, et al. A traditional
cardiovascular diseases. J Cell Biochem. 2018;119:95–104. https://doi. Chinese medicine formula extracts stimulate proliferation and inhibit
org/10.1002/jcb.26169. mineralization of human mesenchymal stem cells in vitro. J Ethnophar-
6. Wu W, Wang Y, Tang Z, Gao Y, Huo Y. Regulatory oversight of cell therapy macol. 2009;125:75–82. https://doi.org/10.1016/j.jep.2009.06.013.
in China: government’s efforts in patient access and therapeutic inno- 25. Choi JH, Lyu SY, Lee HJ, Jung J, Park WB, Kim GJ. Korean mistletoe lectin
vation. Pharmacol Res. 2020;158: 104889. https://doi.org/10.1016/j.phrs. regulates self-renewal of placenta-derived mesenchymal stem cells via
2020.104889. autophagic mechanisms. Cell Prolif. 2012;45:420–9. https://doi.org/10.
7. Udalamaththa VL. Potential role of herbal remedies in stem cell therapy: 1111/j.1365-2184.2012.00839.x.
proliferation and differentiation of human mesenchymal stromal cells. 26. Kim B-S, Kim Y-C, Zadeh H, Park Y-J, Pi S-H, Shin H-S, et al. Effects of the
Stem Cell Res Ther. 2016. https://doi.org/10.1186/s13287-016-0366-4. dichloromethane fraction of Dipsaci Radix on the osteoblastic differ-
8. Jiang L-L, Liu L. Effect of metformin on stem cells: Molecular mecha- entiation of human alveolar bone marrow-derived mesenchymal stem
nism and clinical prospect. World J Stem Cells. 2020;12:1455–73. cells. Biosci Biotechnol Biochem. 2011;75:13–9. https://doi.org/10.1271/
https://doi.org/10.4252/wjsc.v12.i12.1455. bbb.100379.
9. Calabrese EJ. Hormesis and adult adipose-derived stem cells. Pharma- 27. Li G, Zhang X, Zhang J, Chan C, Yew DTW, He M, et al. Ethanol extract of
col Res. 2021;172:105803. https://doi.org/10.1016/j.phrs.2021.105803. Fructus Ligustri Lucidi promotes osteogenesis of mesenchymal stem
10. Haiyan H, Rensong Y, Guoqin J, Xueli Z, Huaying X, Yanwu X. Effect cells. Phytother Res. 2010;24:571–6. https://doi.org/10.1002/ptr.2987.
of astragaloside IV on neural stem cell transplantation in Alzhei- 28. Yoon H-Y, Yun S-I, Kim B-Y, Jin Q, Woo E-R, Jeong S-Y, et al. Poncirin pro-
mer’s disease rat models. Evid Based Complement Alternat Med. motes osteoblast differentiation but inhibits adipocyte differentiation
2016;2016:3106980. https://doi.org/10.1155/2016/3106980. in mesenchymal stem cells. Eur J Pharmacol. 2011;664:54–9. https://doi.
11. Lee JH, Ryu JM, Han Y-S, Zia MF, Kwon HY, Noh H, et al. Fucoidan org/10.1016/j.ejphar.2011.04.047.
improves bioactivity and vasculogenic potential of mesenchymal stem 29. Li X-D, Wang J-S, Chang B, Chen B, Guo C, Hou G-Q, et al. Panax
cells in murine hind limb ischemia associated with chronic kidney notoginseng saponins promotes proliferation and osteogenic dif-
disease. J Mol Cell Cardiol. 2016;97:169–79. https://doi.org/10.1016/j. ferentiation of rat bone marrow stromal cells. J Ethnopharmacol.
yjmcc.2016.05.011. 2011;134:268–74. https://doi.org/10.1016/j.jep.2010.11.075.
12. Zheng P-D, Mungur R, Zhou H-J, Hassan M, Jiang S-N, Zheng J-S. Gink- 30. Chen B, Li X, Liu D, Wang H, Xie P, Liu Z, et al. Canonical Wnt signaling
golide B promotes the proliferation and differentiation of neural stem is required for Panax notoginseng saponin-mediated attenuation of
cells following cerebral ischemia/reperfusion injury, both in vivo and the RANKL/OPG ratio in bone marrow stromal cells during osteogenic
in vitro. Neural Regen Res. 2018;13:1204–11. https://doi.org/10.4103/ differentiation. Phytomedicine. 2012;19:1029–34. https://doi.org/10.
1673-5374.232476. 1016/j.phymed.2012.06.002.
13. Li L, Liu J. Effect of Chinese medicine XIAOJI decoction combined with 31. Yu G-Y, Zheng G-Z, Chang B, Hu Q-X, Lin F-X, Liu D-Z, et al. Naringin
platinum-based chemotherapy and transfusion of cytokine-induced stimulates osteogenic differentiation of rat bone marrow stromal
killer cells in patients with stage III B/IV non-small cell lung cancer. J cells via activation of the notch signaling pathway. Stem Cells Int.
Drug Metab Toxicol. 2015. https://doi.org/10.4172/2157-7609.S10-001. 2016;2016:7130653. https://doi.org/10.1155/2016/7130653.
14. Zhu H, Wang B, Kong L, An T, Li G, Zhou H, et al. Parvifoline AA pro- 32. Zhang P, Peng-Zhang null, Dai K, Yan S, Yan W, Zhang C, et al. Effects of
motes susceptibility of hepatocarcinoma to natural killer cell-mediated naringin on the proliferation and osteogenic differentiation of human
cytolysis by targeting peroxiredoxin. Cell Chem Biol. 2019;26:1122-1132. bone mesenchymal stem cell. Eur J Pharmacol. 2009;607:1–5. https://
e6. https://doi.org/10.1016/j.chembiol.2019.04.003. doi.org/10.1016/j.ejphar.2009.01.035
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 11 of 14
33. Yonezawa T, Lee J-W, Hibino A, Asai M, Hojo H, Cha B-Y, et al. Harmine 51. Zhou Y, Wu Y, Jiang X, Zhang X, Xia L, Lin K, et al. The effect of quercetin
promotes osteoblast differentiation through bone morphogenetic pro- on the osteogenesic differentiation and angiogenic factor expression
tein signaling. Biochem Biophys Res Commun. 2011;409:260–5. https:// of bone marrow-derived mesenchymal stem cells. PLoS ONE. 2015;10:
doi.org/10.1016/j.bbrc.2011.05.001. e0129605. https://doi.org/10.1371/journal.pone.0129605.
34. Jin P, Li M, Xu G, Zhang K, Zheng LI, Zhao J. Role of (-)-epigallocatechin- 52. Geng L, Liu Z, Zhang W, Li W, Wu Z, Wang W, et al. Chemical screen
3-gallate in the osteogenic differentiation of human bone marrow identifies a geroprotective role of quercetin in premature aging. Protein
mesenchymal stem cells: an enhancer or an inducer? Exp Ther Med. Cell. 2019;10:417–35. https://doi.org/10.1007/s13238-018-0567-y.
2015;10:828–34. https://doi.org/10.3892/etm.2015.2579. 53. Kim BS, Kang H-J, Park J-Y, Lee J. Fucoidan promotes osteoblast dif-
35. Park S-J, Lee KW, Lim D-S, Lee S. The sulfated polysaccharide fucoidan ferentiation via JNK- and ERK-dependent BMP2-Smad 1/5/8 signaling in
stimulates osteogenic differentiation of human adipose-derived stem human mesenchymal stem cells. Exp Mol Med. 2015;47: e128. https://
cells. Stem Cells Dev. 2012;21:2204–11. https://doi.org/10.1089/scd. doi.org/10.1038/emm.2014.95.
2011.0521. 54. Ryu BM, Li YX, Kang KH, Kim SK, Kim DG. Floridoside from Laurencia
36. Zhou C, Lin Y. Osteogenic differentiation of adipose-derived stem cells undulata promotes osteogenic differentiation in murine bone marrow
promoted by quercetin. Cell Prolif. 2014;47:124–32. https://doi.org/10. mesenchymal cells. J Funct Foods. 2015;19:505–11.
1111/cpr.12097. 55. Shakibaei M, Shayan P, Busch F, Aldinger C, Buhrmann C, Lueders C,
37. Qiu X-M, Wang L, Gui Y-Y, Xu Y-P, Li D-J. BSNXD modulates mesenchymal et al. Resveratrol mediated modulation of Sirt-1/Runx2 promotes
stem cell differentiation into osteoblasts in a postmenopausal osteo- osteogenic differentiation of mesenchymal stem cells: potential role
porotic mouse model. Int J Clin Exp Pathol. 2015;8:4408–17. of Runx2 deacetylation. PLoS ONE. 2012;7: e35712. https://doi.org/10.
38. Zhang J, Li G, Chan C, Meng C, Lin MC, Chen Y, et al. Flavonoids of 1371/journal.pone.0035712.
herba epimedii regulate osteogenesis of human mesenchymal stem 56. Peltz L, Gomez J, Marquez M, Alencastro F, Atashpanjeh N, Quang T,
cells through BMP and Wnt/beta-catenin signaling pathway. Mol Cell et al. Resveratrol exerts dosage and duration dependent effect on
Endocrinol. 2010;314:70–4. https://doi.org/10.1016/j.mce.2009.08.012. human mesenchymal stem cell development. PLoS ONE. 2012;7:
39. Liao F, Liu Y, Liu H-H, Hu J, Zhao S, Yang S-M. Effect of Angelica sinensis e37162. https://doi.org/10.1371/journal.pone.0037162.
polysaccharide on the osteogenic differentiation of bone marrow mes- 57. Tse GG, Kim BB, McMurtray AM, Nakamoto BK. Case of levodopa toxic-
enchymal stem cells of rats with high glucose levels. Hua Xi Kou Qiang ity from ingestion of Mucuna gigantea. Hawaii J Med Public Health.
Yi Xue Za Zhi. 2019;37:193–9. https://doi.org/10.7518/hxkq.2019.02.012. 2013;72:157–60.
40. Gu Q, Chen C, Zhang Z, Wu Z, Fan X, Zhang Z, et al. Ginkgo biloba 58. Kongros K. The effects of seed extract of Mucuna gigantea on the
extract promotes osteogenic differentiation of human bone marrow expression of neural markers in mesenchymal stem cells. J Med Plants
mesenchymal stem cells in a pathway involving Wnt/β-catenin signal- Res. 2012. https://doi.org/10.5897/JMPR11.1406.
ing. Pharmacol Res. 2015;97:70–8. https://doi.org/10.1016/j.phrs.2015. 59. Ma L, Feng X, Cui B, Law F, Jiang X, Yang L-Y, et al. Human umbilical cord
04.004. Wharton’s Jelly-derived mesenchymal stem cells differentiation into
41. Tao K, Xiao D, Weng J, Xiong A, Kang B, Zeng H. Berberine promotes nerve-like cells. Chin Med J (Engl). 2005;118:1987–93.
bone marrow-derived mesenchymal stem cells osteogenic differen- 60. Hu L, Yu J, Li F, Chen B, Li L, Liu G. Effects of Salvia miltorrhiza in neural
tiation via canonical Wnt/β-catenin signaling pathway. Toxicol Lett. differentiation of rat mesenchymal stem cells with optimized protocol.
2016;240:68–80. https://doi.org/10.1016/j.toxlet.2015.10.007. J Ethnopharmacol. 2011;136:334–40. https://doi.org/10.1016/j.jep.2011.
42. Xu D, Xu L, Zhou C, Lee WYW, Wu T, Cui L, et al. Salvianolic acid B 04.043.
promotes osteogenesis of human mesenchymal stem cells through 61. Si Y-C, Li Q, Xie C-E, Niu X, Xia X-H, Yu C-Y. Chinese herbs and their active
activating ERK signaling pathway. Int J Biochem Cell Biol. 2014;51:1–9. ingredients for activating xue (blood) promote the proliferation and
https://doi.org/10.1016/j.biocel.2014.03.005. differentiation of neural stem cells and mesenchymal stem cells. Chin
43. Cui L, Li T, Liu Y, Zhou L, Li P, Xu B, et al. Salvianolic acid B prevents bone Med. 2014;9:13. https://doi.org/10.1186/1749-8546-9-13.
loss in prednisone-treated rats through stimulation of osteogenesis and 62. Dong L, Wang Y, Lu C, Wang F. Effect of Astragalus mongholicus on
bone marrow angiogenesis. PLoS ONE. 2012;7: e34647. https://doi.org/ inducing differentiations of rat bone marrow-derived mesenchymal
10.1371/journal.pone.0034647. stem cells into neurocyte-like cells. Sichuan Da Xue Xue Bao Yi Xue Ban.
44. Wang L, Zhang Y-G, Wang X-M, Ma L-F, Zhang Y-M. Naringin protects 2007;38:417–20.
human adipose-derived mesenchymal stem cells against hydrogen 63. Dong J, Zhu G, Wang T-C, Shi F-S. Ginsenoside Rg1 promotes neural
peroxide-induced inhibition of osteogenic differentiation. Chem Biol differentiation of mouse adipose-derived stem cells via the miRNA-124
Interact. 2015;242:255–61. https://doi.org/10.1016/j.cbi.2015.10.010. signaling pathway. J Zhejiang Univ Sci B. 2017;18:445–8. https://doi.org/
45. Zhu B, Xue F, Zhang C, Li G. Ginkgolide B promotes osteoblast differen- 10.1631/jzus.B1600355.
tiation via activation of canonical Wnt signalling and alleviates osteo- 64. Wang Q, Zhou L, Guo Y, Liu G, Cheng J, Yu H. Differentiation of human
porosis through a bone anabolic way. J Cell Mol Med. 2019;23:5782–93. adipose-derived stem cells into neuron-like cells by Radix Angelicae
https://doi.org/10.1111/jcmm.14503. Sinensis. Neural Regen Res. 2013;8:3353–8. https://doi.org/10.3969/j.
46. Ying X, Sun L, Chen X, Xu H, Guo X, Chen H, et al. Silibinin promotes issn.1673-5374.2013.35.010.
osteoblast differentiation of human bone marrow stromal cells via 65. Warrier SR, Haridas N, Balasubramanian S, Jalisatgi A, Bhonde R, Dhar-
bone morphogenetic protein signaling. Eur J Pharmacol. 2013;721:225– marajan A. A synthetic formulation, Dhanwantharam kashaya, delays
30. https://doi.org/10.1016/j.ejphar.2013.09.031. senescence in stem cells. Cell Prolif. 2013;46:283–90. https://doi.org/10.
47. Dai J, Li Y, Zhou H, Chen J, Chen M, Xiao Z. Genistein promotion of 1111/cpr.12026.
osteogenic differentiation through BMP2/SMAD5/RUNX2 signaling. Int 66. Widowati W, Sardjono CT, Wijaya L, Laksmitawati DR, Sandra F. Extract of
J Biol Sci. 2013;9:1089–98. https://doi.org/10.7150/ijbs.7367. Curcuma longa L. and (-)-Epigallo Catechin-3-gallate enhanced prolif-
48. Wang J-Y, Chen W-M, Wen C-S, Hung S-C, Chen P-W, Chiu J-H. Du- eration of adipose tissue–derived mesenchymal stem cells (AD-MSCs)
Huo-Ji-Sheng-Tang and its active component Ligusticum chuanxiong and differentiation of AD-MSCs into endothelial progenitor cells. J US
promote osteogenic differentiation and decrease the aging process of China Med Sci. 2012. https://doi.org/10.17265/1548-6648/2012.01.003.
human mesenchymal stem cells. J Ethnopharmacol. 2017;198:64–72. 67. Han Y-S, Lee JH, Jung JS, Noh H, Baek MJ, Ryu JM, et al. Fucoidan
https://doi.org/10.1016/j.jep.2016.12.011. protects mesenchymal stem cells against oxidative stress and enhances
49. Wu Y-Q, Xia L-G, Zhou Y-N, Xu Y-J, Jiang X-Q. Icariin induces osteogenic vascular regeneration in a murine hindlimb ischemia model. Int J
differentiation of bone mesenchymal stem cells in a MAPK-dependent Cardiol. 2015;198:187–95. https://doi.org/10.1016/j.ijcard.2015.06.070.
manner. Cell Prolif. 2015. https://doi.org/10.1111/cpr.12185. 68. Aziz J, Abu Kassim NL, Abu Kasim NH, Haque N, Rahman MT. Carica
50. Zhai Y-K, Guo X-Y, Ge B-F, Zhen P, Ma X-N, Zhou J, et al. Icariin stimulates papaya induces in vitro thrombopoietic cytokines secretion by mesen-
the osteogenic differentiation of rat bone marrow stromal cells via chymal stem cells and haematopoietic cells. BMC Complement Altern
activating the PI3K-AKT-eNOS-NO-cGMP-PKG. Bone. 2014;66:189–98. Med. 2015;15:215. https://doi.org/10.1186/s12906-015-0749-6.
https://doi.org/10.1016/j.bone.2014.06.016. 69. Di Giacomo C, Vanella L, Sorrenti V, Santangelo R, Barbagallo I, Calabrese
G, Genovese C, Mastrojeni S, Ragusa S, Acquaviva R. Effects of Tithonia
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 12 of 14
diversifolia (Hemsl.) A. Gray extract on adipocyte differentiation of 87. Gugliandolo E, D’Amico R, Cordaro M, Fusco R, Siracusa R, Crupi R, et al.
human mesenchymal stem cells. PLOS ONE. 2015;10(4):e0122320. Neuroprotective effect of artesunate in experimental model of trau-
https://doi.org/10.1371/journal.pone.0122320. matic brain injury. Front Neurol. 2018;9:590. https://doi.org/10.3389/
70. Subash-Babu P, Alshatwi AA. Aloe-emodin inhibits adipocyte differen- fneur.2018.00590.
tiation and maturation during in vitro human mesenchymal stem cell 88. Zhang K, Yang Y, Hongfei Ge J, Wang XC, Lei X, Zhong J, Zhang C, Xian
adipogenesis. J Biochem Mol Toxicol. 2012;26:291–300. https://doi.org/ J, Yongling L, Tan L, Feng H. Artesunate promotes the proliferation
10.1002/jbt.21415. of neural stem/progenitor cells and alleviates Ischemia-reperfusion
71. Yan JS, Wei ZJ, Wang B, Song ZL. Interventional effect of Quzhisu on adi- Injury through PI3K/Akt/FOXO-3a/p27kip1 signaling pathway. Aging.
pogenic differentiation of mesenchymal stem cells derived from aplas- 2020;12(9):8029–48. https://doi.org/10.18632/aging.103121.
tic anemia patients. J Clin Rehabilit Tissue Eng Res. 2008;12:4873–6. 89. Li M-Y, Chang C-T, Han Y-T, Liao C-P, Yu J-Y, Wang T-W. Ginkgolide B
72. Xu Y, Wu C, Wu Y, Tong P, Jin H, Yu N, et al. Epimedium-derived flavo- promotes neuronal differentiation through the Wnt/β-catenin pathway
noids modulate the balance between osteogenic differentiation and in neural stem cells of the postnatal mammalian subventricular zone.
adipogenic differentiation in bone marrow stromal cells of ovariecto- Sci Rep. 2018;8:14947. https://doi.org/10.1038/s41598-018-32960-8.
mized rats via Wnt/β-catenin signal pathway activation. Chin J Integr 90. Tiwari SK, Agarwal S, Seth B, Yadav A, Nair S, Bhatnagar P, et al.
Med. 2012;18:909–17. https://doi.org/10.1007/s11655-012-1294-2. Curcumin-loaded nanoparticles potently induce adult neurogenesis
73. Jeong S-G, Oh YS, Joe I-S, Jeong SY, Cho HM, Lee JS, et al. Functional and reverse cognitive deficits in Alzheimer’s disease model via canoni-
restoration of replicative senescent mesenchymal stem cells by the cal Wnt/β-catenin pathway. ACS Nano. 2014;8:76–103. https://doi.org/
brown alga Undaria pinnatifida. Anim Cells Syst (Seoul). 2017;21:108–14. 10.1021/nn405077y.
https://doi.org/10.1080/19768354.2017.1292951. 91. Yang P, Guan Y-Q, Li Y-L, Zhang L, Zhang L, Li L. Icariin promotes cell
74. Liu D, Tang W, Zhang H, Huang H, Zhang Z, Tang D, et al. Icariin protects proliferation and regulates gene expression in human neural stem cells
rabbit BMSCs against OGD-induced apoptosis by inhibiting ERs-medi- in vitro. Mol Med Rep. 2016;14:1316–22. https://doi.org/10.3892/mmr.
ated autophagy via MAPK signaling pathway. Life Sci. 2020;253: 117730. 2016.5377.
https://doi.org/10.1016/j.lfs.2020.117730. 92. Huang J, Cai W, Zhang X, Shen Z. Icariin promotes self-renewal of neural
75. Cruciani S, Santaniello S, Fadda A, Sale L, Sarais G, Sanna D, et al. Extracts stem cells: an involvement of extracellular regulated kinase signaling
from myrtle liqueur processing waste modulate stem cells pluripotency pathway. Chin J Integr Med. 2014;20:107–15. https://doi.org/10.1007/
under stressing conditions. Biomed Res Int. 2019;2019:5641034. https:// s11655-013-1583-7.
doi.org/10.1155/2019/5641034. 93. Lu Q, Zhu H, Liu X, Tang C. Icariin sustains the proliferation and dif-
76. Cruciani S, Garroni G, Ginesu GC, Fadda A, Ventura C, Maioli M. Unravel- ferentiation of Aβ25-35-treated hippocampal neural stem cells via the
ling cellular mechanisms of stem cell senescence: an aid from natural BDNF-TrkB-ERK/Akt signaling pathway. Neurol Res. 2020;42:936–45.
bioactive molecules. Biology (Basel). 2020;9:E57. https://doi.org/10. https://doi.org/10.1080/01616412.2020.1792701.
3390/biology9030057. 94. Guo G, Li B, Wang Y, Shan A, Shen W, Yuan L, et al. Effects of salvia-
77. Lee JH, Jung HK, Han Y-S, Yoon YM, Yun CW, Sun HY, et al. Antioxidant nolic acid B on proliferation, neurite outgrowth and differentiation
effects of Cirsium setidens extract on oxidative stress in human mesen- of neural stem cells derived from the cerebral cortex of embryonic
chymal stem cells. Mol Med Rep. 2016;14:3777–84. https://doi.org/10. mice. Sci China Life Sci. 2010;53:653–62. https://doi.org/10.1007/
3892/mmr.2016.5706. s11427-010-3106-5.
78. Jeong S-H, Lee J-E, Kim B-B, Ko Y, Park J-B. Evaluation of the effects of 95. Zhuang P, Zhang Y, Cui G, Bian Y, Zhang M, Zhang J, et al. Direct stimula-
Cimicifugae Rhizoma on the morphology and viability of mesenchymal tion of adult neural stem/progenitor cells in vitro and neurogenesis
stem cells. Exp Ther Med. 2015;10:629–34. https://doi.org/10.3892/etm. in vivo by salvianolic acid B. PLoS ONE. 2012;7: e35636. https://doi.org/
2015.2578. 10.1371/journal.pone.0035636.
79. Jeong S-H, Lee J-E, Jin S-H, Ko Y, Park J-B. Effects of Asiasari radix on the 96. Yan R, Xu H, Fu X. Salidroside protects hypoxia-induced injury by up-
morphology and viability of mesenchymal stem cells derived from the regulation of miR-210 in rat neural stem cells. Biomed Pharmacother.
gingiva. Mol Med Rep. 2014;10:3315–9. https://doi.org/10.3892/mmr. 2018;103:1490–7. https://doi.org/10.1016/j.biopha.2018.04.184.
2014.2607. 97. Chai Y-S, Hu J, Lei F, Wang Y-G, Yuan Z-Y, Lu X, et al. Effect of berberine
80. Wang J, Hu J, Chen X, Lei X, Feng H, Wan F, et al. Traditional Chinese on cell cycle arrest and cell survival during cerebral ischemia and
medicine monomers: novel strategy for endogenous neural stem cells reperfusion and correlations with p53/cyclin D1 and PI3K/Akt. Eur J
activation after stroke. Front Cell Neurosci. 2021;15: 628115. https://doi. Pharmacol. 2013;708:44–55. https://doi.org/10.1016/j.ejphar.2013.02.
org/10.3389/fncel.2021.628115. 041.
81. Grochowski C, Radzikowska E, Maciejewski R. Neural stem cell therapy- 98. Chen D-F, Meng L-J, Du S-H, Zhang H-L, Li H, Zhou J-H, et al. (+)-Cho-
brief review. Clin Neurol Neurosurg. 2018;173:8–14. https://doi.org/10. lesten-3-one induces differentiation of neural stem cells into dopamin-
1016/j.clineuro.2018.07.013. ergic neurons through BMP signaling. Neurosci Res. 2010;68:176–84.
82. Qin W, Chen S, Yang S, Qian X, Chuanshan X, Cai J. The effect of https://doi.org/10.1016/j.neures.2010.07.2043.
traditional Chinese medicine on neural stem cell proliferation and 99. Huang F, Lan Y, Qin L, Dong H, Shi H, Wu H, et al. Astragaloside IV
differentiation. Aging Dis. 2017;8(6):792. https://doi.org/10.14336/AD. promotes adult neurogenesis in hippocampal dentate gyrus of mouse
2017.0428. through CXCL1/CXCR2 signaling. Molecules. 2018;23:E2178. https://doi.
83. Gao J, Wan F, Tian M, Li Y, Li Y, Li Q, et al. Effects of ginsenoside-Rg1 on org/10.3390/molecules23092178.
the proliferation and glial-like directed differentiation of embryonic 100. Si Y-C, Zhang J-P, Xie C-E, Zhang L-J, Jiang X-N. Effects of Panax notogin-
rat cortical neural stem cells in vitro. Mol Med Rep. 2017;16:8875–81. seng saponins on proliferation and differentiation of rat hippocampal
https://doi.org/10.3892/mmr.2017.7737. neural stem cells. Am J Chin Med. 2011;39:999–1013. https://doi.org/10.
84. Lin T, Liu Y, Shi M, Liu X, Li L, Liu Y, et al. Promotive effect of ginsenoside 1142/S0192415X11009366.
Rd on proliferation of neural stem cells in vivo and in vitro. J Ethnophar- 101. Tian Y, Liu Y, Chen X, Zhang H, Shi Q, Zhang J, et al. Tetramethylpyrazine
macol. 2012;142:754–61. https://doi.org/10.1016/j.jep.2012.05.057. promotes proliferation and differentiation of neural stem cells from rat
85. Cheng W, Yu P, Wang L, Shen C, Song X, Chen J, et al. Sonic hedgehog brain in hypoxic condition via mitogen-activated protein kinases path-
signaling mediates resveratrol to increase proliferation of neural stem way in vitro. Neurosci Lett. 2010;474:26–31. https://doi.org/10.1016/j.
cells after oxygen-glucose deprivation/reoxygenation injury in vitro. neulet.2010.02.066.
Cell Physiol Biochem. 2015;35:2019–32. https://doi.org/10.1159/00037 102. Zhuang P-W, Cui G-Z, Zhang Y-J, Zhang M-X, Guo H, Zhang J-B, et al.
4009. Baicalin regulates neuronal fate decision in neural stem/progenitor cells
86. Shen C, Cheng W, Yu P, Wang L, Zhou L, Zeng L, et al. Resveratrol and stimulates hippocampal neurogenesis in adult rats. CNS Neurosci
pretreatment attenuates injury and promotes proliferation of neural Ther. 2013;19:154–62. https://doi.org/10.1111/cns.12050.
stem cells following oxygen-glucose deprivation/reoxygenation by 103. Li Y, Zhuang P, Shen B, Zhang Y, Shen J. Baicalin promotes neuronal
upregulating the expression of Nrf2, HO-1 and NQO1 in vitro. Mol Med differentiation of neural stem/progenitor cells through modulating
Rep. 2016;14:3646–54. https://doi.org/10.3892/mmr.2016.5670.
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 13 of 14
p-stat3 and bHLH family protein expression. Brain Res. 2012;1429:36– 121. Nembo EN, Atsamo AD, Nguelefack TB, Kamanyi A, Hescheler J,
42. https://doi.org/10.1016/j.brainres.2011.10.030. Nguemo F. In vitro chronotropic effects of Erythrina senegalensis DC
104. Sun J, Bi Y, Guo L, Qi X, Zhang J, Li G, et al. Buyang Huanwu Decoction (Fabaceae) aqueous extract on mouse heart slice and pluripotent stem
promotes growth and differentiation of neural progenitor cells: using a cell-derived cardiomyocytes. J Ethnopharmacol. 2015;165:163–72.
serum pharmacological method. J Ethnopharmacol. 2007;113:199–203. https://doi.org/10.1016/j.jep.2015.02.002.
https://doi.org/10.1016/j.jep.2007.05.018. 122. You J, Roh K-B, Li Z, Liu G, Tang J, Shin S, et al. The antiaging properties
105. Sun J-H, Gao Y-M, Yang L, Wang X, Bao L-H, Liu W-J, et al. Effects of of andrographis paniculata by activation epidermal cell stemness. Mol-
Buyang Huanwu Decoction on neurite outgrowth and differentiation ecules. 2015;20:17557–69. https://doi.org/10.3390/molecules200917
of neuroepithelial stem cells. Chin J Physiol. 2007;50:151–6. 557.
106. Wu L, Ran C, Liu S, Liao L, Chen Y, Guo H, et al. Jiaweisinisan facilitates 123. Lee J, Shin Y-K, Song J-Y, Lee K-W. Protective mechanism of morin
neurogenesis in the hippocampus after stress damage. Neural Regen against ultraviolet B-induced cellular senescence in human keratino-
Res. 2013;8:1091–102. https://doi.org/10.3969/j.issn.1673-5374.2013.12. cyte stem cells. Int J Radiat Biol. 2014;90:20–8. https://doi.org/10.3109/
004. 09553002.2013.835502.
107. Liang G, Zhang Y. Embryonic stem cell and induced pluripotent stem 124. Lee J, Cho JY, Lee SY, Lee K-W, Lee J, Song J-Y. Vanillin protects human
cell: an epigenetic perspective. Cell Res. 2013;23:49–69. https://doi.org/ keratinocyte stem cells against ultraviolet B irradiation. Food Chem
10.1038/cr.2012.175. Toxicol. 2014;63:30–7. https://doi.org/10.1016/j.fct.2013.10.031.
108. Pouton CW, Haynes JM. Embryonic stem cells as a source of models for 125. Lee J, Oh SW, Shin SW, Lee K-W, Cho J-Y, Lee J. Zingerone protects
drug discovery. Nat Rev Drug Discov. 2007;6:605–16. https://doi.org/10. keratinocyte stem cells from UVB-induced damage. Chem Biol Interact.
1038/nrd2194. 2018;279:27–33. https://doi.org/10.1016/j.cbi.2017.11.004.
109. Li Y, Liu Z, Li J, Wang M. Traditional Chinese medicine, Kami-Shoyo-San 126. Chen J, Wang X, Zhu J, Shang Y, Guo X, Sun J. Effects of Ginkgo biloba
protects ketamine-induced neurotoxicity in human embryonic stem extract on number and activity of endothelial progenitor cells from
cell-differentiated neurons through activation of brain-derived neuro- peripheral blood. J Cardiovasc Pharmacol. 2004;43:347–52. https://doi.
trophic factor. NeuroReport. 2019;30:1102–9. org/10.1097/00005344-200403000-00004.
110. Kim KM, Heo DR, Lee JY, Seo C-S, Chung S-K. High-efficiency generation 127. Dong XX, Hui ZJ, Xiang WX, Rong ZF, Jian S, Zhu CJ. Ginkgo biloba
of induced pluripotent stem cells from human foreskin fibroblast cells extract reduces endothelial progenitor-cell senescence through aug-
using the Sagunja-tang herbal formula. BMC Complement Altern Med. mentation of telomerase activity. J Cardiovasc Pharmacol. 2007;49:111–
2017;17:529. https://doi.org/10.1097/WNR.0000000000001328. 5. https://doi.org/10.1097/FJC.0b013e31802ef519.
111. Kim A, Lee S-Y, Seo C-S, Chung S-K. Prunellae spica extract suppresses 128. Weibo Y, Qin X, Jin Y, Li Y, Santiskulvong C, Victor V, Zeng G, Zhang Z,
teratoma formation of pluripotent stem cells through p53-mediated Chow M, Rao J. Tianshengyuan-1 (TSY-1) regulates cellular Telomerase
apoptosis. Nutrients. 2020;12:E721. https://doi.org/10.3390/nu120 activity by methylation of TERT promoter. Oncotarget. 2016;8(5):7977–
30721. 88. https://doi.org/10.18632/oncotarget.13939.
112. Kim A, Lee S-Y, Seo C-S, Chung S-K. Ethanol extract of Magnoliae cortex 129. Wang Z, Cao YJ. Adoptive cell therapy targeting neoantigens: a frontier
(EEMC) limits teratoma formation of pluripotent stem cells by selective for cancer research. Front Immunol. 2020;11:176. https://doi.org/10.
elimination of undifferentiated cells through the p53-dependent mito- 3389/fimmu.2020.00176.
chondrial apoptotic pathway. Phytomedicine. 2020;69: 153198. https:// 130. Chen N, Xu Y, Mou J, Rao Q, Xing H, Tian Z, et al. Targeting of IL-10R
doi.org/10.1016/j.phymed.2020.153198. on acute myeloid leukemia blasts with chimeric antigen receptor-
113. Shu T, Pang M, Rong L, Zhou W, Wang J, Liu C, et al. Effects of salvia expressing T cells. Blood Cancer J. 2021;11:144. https://doi.org/10.1038/
miltiorrhiza on neural differentiation of induced pluripotent stem cells. s41408-021-00536-x.
J Ethnopharmacol. 2014;153:233–41. https://doi.org/10.1016/j.jep.2014. 131. Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. “Off-the-shelf” allo-
02.028. geneic CAR T cells: development and challenges. Nat Rev Drug Discov.
114. Chang C-Y, Chen S-M, Lu H-E, Lai S-M, Lai P-S, Shen P-W, et al. 2020;19:185–99. https://doi.org/10.1038/s41573-019-0051-2.
N-butylidenephthalide attenuates Alzheimer’s disease-like cytopathy 132. Wilkins O, Keeler AM, Flotte TR. CAR T-cell therapy: progress and pros-
in down syndrome induced pluripotent stem cell-derived neurons. Sci pects. Hum Gene Ther Methods. 2017;28:61–6. https://doi.org/10.1089/
Rep. 2015;5:8744. https://doi.org/10.1038/srep08744. hgtb.2016.153.
115. Pei-Chun W, Ming-Ji Fann T, Tran T, Chen SC, Devina T, Cheng IHJ, Lien 133. Larson RC, Maus MV. Recent advances and discoveries in the mecha-
CC, Kao LS, Wang SJ, Fuh JL, Tzeng TT, Huang CY, Shiao YJ, Wong YH. nisms and functions of CAR T cells. Nat Rev Cancer. 2021;21:145–61.
Assessing the therapeutic potential of Graptopetalum paraguayense on https://doi.org/10.1038/s41568-020-00323-z.
Alzheimer’s disease using patient iPSC-derived neurons. Sci Rep. 2019. 134. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev
https://doi.org/10.1038/s41598-019-55614-9. Clin Oncol. 2019;16:372–85. https://doi.org/10.1038/s41571-019-0184-6.
116. Wei W, Liu Y, Zhang Q, Wang Y, Zhang X, Zhang H. Danshen-enhanced 135. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome
cardioprotective effect of cardioplegia on ischemia reperfusion injury the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol.
in a human-induced pluripotent stem cell-derived cardiomyocytes 2020;17:147–67. https://doi.org/10.1038/s41571-019-0297-y.
model. Artif Organs. 2017;41:452–60. https://doi.org/10.1111/aor.12801. 136. Lesch S, Benmebarek M-R, Cadilha BL, Stoiber S, Subklewe M, Endres
117. Pahlavan S, Tousi MS, Ayyari M, Alirezalu A, Ansari H, Saric T, et al. Effects S, et al. Determinants of response and resistance to CAR T cell therapy.
of hawthorn (Crataegus pentagyna) leaf extract on electrophysiologic Semin Cancer Biol. 2020;65:80–90. https://doi.org/10.1016/j.semcancer.
properties of cardiomyocytes derived from human cardiac arrhythmia- 2019.11.004.
specific induced pluripotent stem cells. FASEB J. 2018;32:1440–51. 137. Chen X, Cao Z, Zhang Y, Li J, Wang S, Du J, et al. Fuzheng Qingjie
https://doi.org/10.1096/fj.201700494RR. Granules inhibit growth of hepatoma cells via inducing mitochondria-
118. Zhang M, Wu H, Guo F, Yu Y, Wei J, Geng Y, et al. Identification of active mediated apoptosis and enhancing immune function. Integr Cancer
components in Yixinshu Capsule with protective effects against myo- Ther. 2017;16:329–38. https://doi.org/10.1177/1534735416654761.
cardial dysfunction on human induced pluripotent stem cell-derived 138. Chen X-Z, Cao Z-Y, Zhang Y-Q, Li J-N, Liao L-M, Du J. Fuzheng Qingjie
cardiomyocytes by an integrative approach. Mol Biosyst. 2017;13:1469– granules potentiate the anticancer effect of cyclophosphamide
80. https://doi.org/10.1039/c6mb00813e. by regulating cellular immune function and inducing apoptosis in
119. Yu Y, Sun S, Wang S, Zhang Q, Li M, Lan F, et al. Liensinine- and neferine- Hepatoma 22 tumor-bearing mice. Oncol Lett. 2017;13:3261–8. https://
induced cardiotoxicity in primary neonatal rat cardiomyocytes and doi.org/10.3892/ol.2017.5849.
human-induced pluripotent stem cell-derived cardiomyocytes. Int J 139. Liu S, Wang X-M, Yang G-W. Action mechanism of fuzheng fangai pill
Mol Sci. 2016;17:E186. https://doi.org/10.3390/ijms17020186. combined with cyclophosphamide on tumor metastasis and growth.
120. Lu J, Wei H, Wu J, Jamil MFA, Tan ML, Adenan MI, et al. Evaluation of the Evid Based Complement Alternat Med. 2014;2014: 494528. https://doi.
cardiotoxicity of mitragynine and its analogues using human induced org/10.1155/2014/494528.
pluripotent stem cell-derived cardiomyocytes. PLoS ONE. 2014;9: 140. Zhang A, Yang X, Li Q, Yang Y, Zhao G, Wang B, et al. Immunostimula-
e115648. https://doi.org/10.1371/journal.pone.0115648. tory activity of water-extractable polysaccharides from Cistanche
Li et al. Stem Cell Research & Therapy (2022) 13:472 Page 14 of 14
deserticola as a plant adjuvant in vitro and in vivo. PLoS ONE. 2018;13: 158. Cai Y, Xiong S, Zheng Y, Luo F, Jiang P, Chu Y. Trichosanthin enhances
e0191356. https://doi.org/10.1371/journal.pone.0191356. anti-tumor immune response in a murine Lewis lung cancer model by
141. Wang C, Feng L, Jiayan S, Cui L, Liu D, Yan J, Ding C, Tan X, Jia X. Polysac- boosting the interaction between TSLC1 and CRTAM. Cell Mol Immu-
charides from Epimedium koreanum Nakai with immunomodulatory nol. 2011;8:359–67. https://doi.org/10.1038/cmi.2011.12.
activity and inhibitory effect on tumor growth in LLC-bearing mice. J 159. Xu R, Lin L, Li Y, Li Y. ShenQi FuZheng Injection combined with chemo-
Ethnopharmacol. 2017;207:8–18. https://doi.org/10.1016/j.jep.2017.06. therapy in the treatment of colorectal cancer: a meta-analysis. PLoS
014. ONE. 2017;12: e0185254. https://doi.org/10.1371/journal.pone.0185254.
142. Ayeka PA, Bian YH, Githaiga PM, Zhao Y. The immunomodulatory 160. Cao GW, Yang WG, Du P. Observation of the effects of LAK/IL-2 therapy
activities of licorice polysaccharides (Glycyrrhiza uralensis Fisch.) in CT 26 combining with Lycium barbarum polysaccharides in the treatment of
tumor-bearing mice. BMC Complementary Altern Med. 2017. https:// 75 cancer patients. Zhonghua Zhong Liu Za Zhi. 1994;16:428–31.
doi.org/10.1186/s12906-017-2030-7. 161. Saleh MH, Rashedi I, Keating A. Immunomodulatory Properties of
143. Guo A, He D, Xu H-B, Geng C-A, Zhao J. Promotion of regulatory T cell Coriolus versicolor: The role of polysaccharopeptide. Front Immunol.
induction by immunomodulatory herbal medicine licorice and its two 2017;8:1087. https://doi.org/10.3389/fimmu.2017.01087.
constituents. Sci Rep. 2015;5:14046. https://doi.org/10.1038/srep14046. 162. Bao YX, Wong CK, Leung SF, Chan ATC, Li PW, Wong ELY, et al. Clini-
144. Wang Q, Chen D-Y. Effect of Aidi injection on peripheral blood expres- cal studies of immunomodulatory activities of Yunzhi-Danshen in
sion of Th1/Th2 transcription factors and cytokines in patients with patients with nasopharyngeal carcinoma. J Altern Complement Med.
esophageal squamous cell carcinoma during radiotherapy. Zhongguo 2006;12:771–6. https://doi.org/10.1089/acm.2006.12.771.
Zhong Xi Yi Jie He Za Zhi. 2009;29:394–7. 163. Liu J, Wang Y, Qiu Z, Lv G, Huang X, Lin H, et al. Impact of TCM on
145. Zhao H, Li Y, Wang Y, Zhang J, Ouyang X, Peng R, et al. Antitumor and tumor-infiltrating myeloid precursors in the tumor microenvironment.
immunostimulatory activity of a polysaccharide-protein complex from Front Cell Dev Biol. 2021;9: 635122. https://doi.org/10.3389/fcell.2021.
Scolopendra subspinipes mutilans L. Koch in tumor-bearing mice. Food 635122.
Chem Toxicol. 2012;50:2648–55. https://doi.org/10.1016/j.fct.2012.05. 164. Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for
018. cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12:269–
146. Ma W, Liu R, Qi J, Zhang Y. Extracts of centipede Scolopendra subspinipes 81. https://doi.org/10.1038/nri3191.
mutilans induce cell cycle arrest and apoptosis in A375 human mela- 165. Qi Y, Gao F, Hou L, Wan C. Anti-inflammatory and immunostimulatory
noma cells. Oncol Lett. 2014;8:414–20. https://doi.org/10.3892/ol.2014. activities of Astragalosides. Am J Chin Med. 2017;45:1157–67. https://
2139. doi.org/10.1142/S0192415X1750063X.
147. Ding D, Guo Y-R, Wu R-L, Qi W-Y, Xu H-M. Two new isoquinoline alka- 166. Zhang L, Wei W. Anti-inflammatory and immunoregulatory effects of
loids from Scolopendra subspinipes mutilans induce cell cycle arrest and paeoniflorin and total glucosides of paeony. Pharmacol Ther. 2020;207:
apoptosis in human glioma cancer U87 cells. Fitoterapia. 2016;110:103– 107452. https://doi.org/10.1016/j.pharmthera.2019.107452.
9. https://doi.org/10.1016/j.fitote.2016.03.004. 167. Yang R, Yuan B-C, Ma Y-S, Zhou S, Liu Y. The anti-inflammatory activity of
148. Li W, Yang Y, Ouyang Z, Zhang Q, Wang L, Tao F, et al. Xiao-Ai-Ping, a licorice, a widely used Chinese herb. Pharm Biol. 2017;55:5–18. https://
TCM Injection, enhances the Antigrowth effects of cisplatin on lewis doi.org/10.1080/13880209.2016.1225775.
lung cancer cells through promoting the infiltration and function 168. Dai Y, Qiang W, Gui Y, Tan X, Pei T, Lin K, et al. A large-scale transcrip-
of CD8(+) T lymphocytes. Evid Based Complement Alternat Med. tional study reveals inhibition of COVID-19 related cytokine storm by
2013;2013: 879512. https://doi.org/10.1155/2013/879512. traditional Chinese medicines. Sci Bull (Beijing). 2021;66:884–8. https://
149. Deng X, Luo S, Luo X, Hu M, Ma F, Wang Y, et al. Polysaccharides doi.org/10.1016/j.scib.2021.01.005.
from Chinese Herbal Lycium barbarum Induced systemic and local
immune responses in H22 tumor-bearing mice. J Immunol Res.
2018;2018:3431782. https://doi.org/10.1155/2018/3431782. Publisher’s Note
150. Wang W, Liu M, Wang Y, Yang T, Li D, Ding F, et al. Lycium barbarum Springer Nature remains neutral with regard to jurisdictional claims in pub-
polysaccharide promotes maturation of dendritic cell via notch signal- lished maps and institutional affiliations.
ing and strengthens dendritic cell mediated T lymphocyte cytotoxicity
on colon cancer cell CT26-WT. Evid Based Complement Alternat Med.
2018;2018:2305683. https://doi.org/10.1155/2018/2305683.
151. Bo R, Sun Y, Zhou S, Ou N, Gu P, Liu Z, et al. Simple nanoliposomes
encapsulating Lycium barbarum polysaccharides as adjuvants
improve humoral and cellular immunity in mice. Int J Nanomedicine.
2017;12:6289–301. https://doi.org/10.2147/IJN.S136820.
152. Hsieh C-C, Lin W-C, Lee M-R, Hsu S-L, Liu H-S, Kao S-T, et al. Dang-Gui-
Bu-Xai-Tang modulated the immunity of tumor bearing mice. Immu-
nopharmacol Immunotoxicol. 2003;25:259–71. https://doi.org/10.1081/
iph-120020474.
153. Gupta S, Zhang D, Yi J, Shao J. Anticancer activities of Oldenlandia dif-
fusa. J Herb Pharmacother. 2004;4:21–33.
154. Yadav SK, Lee SC. Evidence for Oldenlandia diffusa-evoked cancer cell
apoptosis through superoxide burst and caspase activation. Zhong Xi
Ready to submit your research ? Choose BMC and benefit from:
Yi Jie He Xue Bao. 2006;4:485–9. https://doi.org/10.3736/jcim20060509.
155. Willimott S, Barker J, Jones LA, Opara EI. Apoptotic effect of Oldenlandia
• fast, convenient online submission
diffusa on the leukaemic cell line HL60 and human lymphocytes. J
Ethnopharmacol. 2007;114:290–9. https://doi.org/10.1016/j.jep.2007.08. • thorough peer review by experienced researchers in your field
030. • rapid publication on acceptance
156. Chang J-M, Hung L-M, Chyan Y-J, Cheng C-M, Wu R-Y. Carthamus tincto-
• support for research data, including large and complex data types
rius enhances the antitumor activity of dendritic cell vaccines via polari-
zation toward Th1 cytokines and increase of cytotoxic T lymphocytes. • gold Open Access which fosters wider collaboration and increased citations
Evid Based Complement Alternat Med. 2011;2011: 274858. https://doi. • maximum visibility for your research: over 100M website views per year
org/10.1093/ecam/nen068.
157. Ma Y-H, Cheng W-Z, Gong F, Ma A-L, Yu Q-W, Zhang J-Y, et al. Active Chi- At BMC, research is always in progress.
nese mistletoe lectin-55 enhances colon cancer surveillance through
regulating innate and adaptive immune responses. World J Gastroen- Learn more biomedcentral.com/submissions
terol. 2008;14:5274–81. https://doi.org/10.3748/wjg.14.5274.