Immunology Draft
Immunology Draft
MECHANISMS OF IMMUNE
PROTECTION AND
PATHOGENESIS
Immunology is the study of the immune system, which plays a crucial role in protecting
the body from infections, diseases, and harmful substances. The immune system is a highly
complex network of cells, tissues, organs, and molecules that work together to defend the body
from pathogens (microorganisms like bacteria, viruses, fungi, and parasites) while maintaining
tolerance to the body’s own cells. The immune system consists of two primary components
namely the innate immune system and the adaptive immune system. The adaptive immune
system is further subdivided into two branches called as humoral immunity, mediated by B
cells and antibodies, and cell-mediated immunity, driven by T cells. These systems work
collaboratively to protect the body from invaders.
A significant feature of the immune system is its ability to differentiate between the
body’s own cells and foreign substances. During early development, immune cells that bind to
self-antigens are eliminated to prevent autoimmunity. On the other hand, immune cells that
recognize and bind to foreign antigens are activated, undergo clonal selection, and proliferate to
mount a defense against the pathogen.
History of Immunology:
The origins of immunology trace back thousands of years, where foundational practices
and observations began shaping this essential scientific field. Ancient cultures in regions like
China and India developed early forms of vaccination by introducing small amounts of infectious
material into individuals to safeguard against illnesses such as smallpox. In the late 1700s,
Edward Jenner revolutionized immunology by showing that cowpox inoculation could
successfully prevent smallpox, marking the advent of modern vaccination techniques. Building
upon Jenner's work, Louis Pasteur made significant strides in the 19th century by creating
vaccines for diseases like rabies and anthrax. He also established the germ theory, which
emphasized the role of microorganisms in infectious diseases. The discovery of antibodies by
Emil von Behring and Shibasaburo Kitasato further illuminated the concept of humoral
immunity, while Ilya Metchnikoff's study of phagocytosis offered insights into cellular
immunity. The 20th century saw monumental progress in immunology, including the
identification of the major histocompatibility complex (MHC), the introduction of the clonal
selection theory by Frank Macfarlane Burnet, and the emergence of immunosuppressive
treatments that transformed organ transplantation. In contemporary times, immunology continues
to advance through innovations like monoclonal antibodies, gene editing, and immunotherapy,
addressing a wide spectrum of diseases, including infectious illnesses and cancer. This evolution
showcases the remarkable progression of immunology from ancient practices to an advanced,
specialized scientific discipline central to modern healthcare and medicine.
ancient civilizations such as China and India. Methods like variolation involved the
intentional exposure to small amounts of infectious material to provide protection against
diseases such as smallpox.
1796 – Edward Jenner: Edward Jenner developed the first modern vaccine by
demonstrating that cowpox inoculation could prevent smallpox, marking the birth of
vaccination and modern immunology.
1860s – Louis Pasteur: Louis Pasteur established the germ theory of disease, proving
microorganisms cause infections. He also developed vaccines for rabies and anthrax,
building the foundation for microbiological immunology.
1890 – Emil von Behring and Shibasaburo Kitasato: Discovered antibodies,
introducing the concept of humoral immunity, which involves the production of specific
molecules to neutralize pathogens.
1891 – Ilya Metchnikoff: Metchnikoff identified phagocytosis, demonstrating how cells
like macrophages engulf and destroy pathogens, forming the basis of cellular immunity.
1930s – Discovery of Complement System: Advances in understanding the complement
system, which enhances immune responses, were made during this era.
1958 – Clonal Selection Theory: Frank Macfarlane Burnet proposed the clonal
selection theory, explaining how immune cells are selected to respond to specific
antigens and proliferate upon activation.
1960s – Major Histocompatibility Complex (MHC): MHC molecules were identified,
revealing their essential role in antigen presentation and immune system regulation.
1970s – Monoclonal Antibodies: Georges Köhler and César Milstein pioneered
the application of CRISPR-Cas9 for manipulating immune system components and the
advent of mRNA vaccines, which proved pivotal in combating COVID-19.
The immune system consists of various organs and tissues spread throughout the body. These
immune organs are divided into two main categories:
A. Primary lymphoid organs: These are where lymphocytes (a type of white blood cell) develop
and mature.
B. Secondary lymphoid organs: These trap antigens (foreign substances) from the tissues and
blood, and are where lymphocytes actually interact with antigens.
All these organs are connected by the lymphatic system and blood vessels, forming a functional
unit. The immune system also includes different types of white blood cells, such as lymphocytes (T-
cells, B-cells, and natural killer cells), neutrophils, monocytes, and macrophages. These cells play a key
role in defending the body against foreign elements.
Of all these cell types, only lymphocytes exhibit diversity, specificity, memory, and the ability
to distinguish self from non-self, which are key features of adaptive immune responses. The other white
blood cells support adaptive immunity by activating lymphocytes, enhancing the removal of antigens
through phagocytosis, or secreting immune-effector molecules. Certain white blood cells release
cytokines, which regulate immune responses. Other crucial proteins in the immune system are
antibodies produced by B-lymphocytes and complement proteins, which are activated by antibodies.
MHC Class I proteins are expressed on most nucleated cells and present antigens from
intracellular pathogens to cytotoxic T cells for cell killing. On the other hand, MHC Class II proteins
are restricted to antigen-presenting cells and present antigens from extracellular pathogens to helper
T cells for immune regulation and activation.
When a naive T-cell encounters an antigen presented with an MHC molecule, it proliferates and
differentiates into memory T-cells and various effector T-cells. T-cells are divided into three types: T
helper (Th), T cytotoxic (Tc), and T suppressor (Ts) cells. Th and Tc cells are distinguished by the
presence of CD4 and CD8 membrane glycoproteins, respectively. T cells with CD4 are Th cells, while
those with CD8 are Tc cells. Th cells recognize antigens presented by MHC class II molecules and,
upon activation, secrete growth factors called cytokines. These cytokines play a crucial role in
activating B-cells, Tc cells, macrophages, and other immune cells. The specific cytokine patterns
produced by activated Th cells lead to different immune responses. Th-derived cytokines help Tc cells
recognize antigens presented by MHC class I molecules, leading to the proliferation and differentiation
of Tc cells into Cytotoxic T-lymphocytes (CTLs). Tc cells, unlike helper T-Cells, do not induce
cytokine secretion but exhibit cell-killing activity. CTLs are essential in monitoring and eliminating
cells displaying antigens, such as tumor cells, virus-infected cells, and foreign tissue graft cells. CTLs
target altered self-cells presented by MHC class I molecules.
There are special NK cells known as NK1-T cells, which share features of both T-lymphocytes
and NK cells. These cells have T-cell receptors (TCRs) that interact with CD1 molecules, unlike
normal TCRs that interact with class I or II MHC molecules. Like NK cells, they have CD16 and other
NK receptors that enable them to kill cells.
NK1-T cells rapidly secrete large amounts of cytokines, supporting antibody production by B-
cells, inflammation, and the development and expansion of cytotoxic T-cells. Some immunologists see
these cells as a rapid response system providing early help while conventional T helper (Th) responses
develop.
Mononuclear phagocytes:
Mononuclear phagocytes include immune cells such as monocytes, which circulate in the blood,
and macrophages, which are found in tissues. During hematopoiesis in the bone marrow, granulocyte-
monocyte progenitor cells differentiate into promonocytes. These promonocytes then leave the bone
marrow, enter the blood, and further differentiate into mature monocytes.
Monocytes circulate in the bloodstream for about 8 hours, during which they enlarge. They then
migrate into tissues and differentiate into specific tissue macrophages or dendritic cells. This
differentiation involves several changes:
The cell enlarges five to tenfold.
Its intracellular organelles increase in number and complexity.
It gains increased phagocytic ability and produces higher levels of hydrolytic enzymes.
It begins to secrete various soluble factors.
Structure of Monocyte
https://biologynotesonline.com/wp-content/uploads/2024/04/image-1475.png
Macrophages are spread throughout the body. Some become fixed macrophages residing in
specific tissues, while others remain motile, called free or wandering macrophages. Free macrophages
move throughout the tissues using amoeboid movement. Macrophage-like cells perform various
functions in different tissues and are named based on their location:
1) Alveolar macrophages in the lungs.
2) Histiocytes in connective tissues.
3) Kupffer cells in the liver.
4) Mesangial cells in the kidneys.
5) Microglial cells in the brain.
6) Osteoclasts in bones.
Structure of Macrophage
Image Source: https://biologynotesonline.com/wp-content/uploads/2024/04/image-1475.png
Macrophages are usually in a resting phase but can be activated by several immune responses.
Initial activation often occurs through the phagocytosis of certain antigens. However, macrophage
activity can be further enhanced by cytokines secreted by activated T helper (TH) cells, inflammatory
response mediators, and bacterial cell wall components. Interferon-gamma, secreted by activated TH
cells, is one of the most potent macrophage activators.
Activated macrophages are more effective at eliminating potential pathogens than resting
macrophages. They exhibit greater phagocytic activity, an increased ability to kill ingested microbes,
increased secretion of inflammatory mediators, and an enhanced ability to activate T cells.
Additionally, activated macrophages secrete various cytotoxic proteins that help eliminate a broad
range of pathogens, including virus-infected cells, tumor cells, and intracellular bacteria. Activated
macrophages also express higher levels of class II MHC molecules, allowing them to function more
effectively as antigen-presenting cells.
Some functions of macrophages include:
✿ Phagocytosis: Engulfing bacteria, viruses, and other foreign particles. Macrophages have Fc
receptors that interact with the Fc component of IgG, facilitating the ingestion of opsonized
organisms. They also have receptors for C3b, another important opsonin. After ingestion, the
phagosome fuses with a lysosome, and the microbe within is killed by reactive oxygen, reactive
nitrogen compounds, and lysosomal enzymes.
✿ Antimicrobial and cytotoxic activities: Includes both oxygen-dependent and oxygen-
independent killing.
✿ Antigen processing: After ingesting and degrading foreign materials, antigen fragments are
presented on the macrophage cell surface with class II MHC proteins for interaction with CD4+
helper T cells. The degradation of the foreign protein stops following the association with class
II MHC proteins in the cytoplasm. The complex is then transported to the cell surface by
transporter proteins.
✿ Secretion of growth factors: Important for the development of an immune response, including
cytokines like interleukin 1 (IL-1), TNF-α, and interleukin 6 (IL-6), which promote
inflammatory responses, complement proteins, hydrolytic enzymes, and a cascade of Tumor
Necrosis Factors (TNF-α) that induce and kill tumor cells and promote hematopoiesis.
Macrophage function
Image source: https://cdn.britannica.com/89/20889-050-9137C72F/Macrophages-components-immune-system-bacteria-particles.jpg
Granulocytes
These are a type of white blood cell (leukocytes). They are classified by their cellular shapes
and how their cytoplasm stains. This group includes neutrophils, eosinophils, basophils, and mast cells.
All granulocytes have distinctive multilobed nuclei, making them easy to identify compared to
lymphocytes, which have round nuclei. The cytoplasm of granulocytes contains numerous granules that
release proteins when they encounter pathogens. These proteins have various roles, including directly
damaging pathogens, regulating other white blood cells, and helping to remodel tissues at infection
sites.
1) Neutrophils have a multilobed nucleus and granulated cytoplasm that stains with both acid and
basic dyes, earning them the name polymorphonuclear leukocytes (PMNs).
2) Eosinophils have a bilobed nucleus and granulated cytoplasm that stains red with the acid dye
eosin.
3) Basophils have a lobed nucleus and granulated cytoplasm that stains blue with the basic dye
methylene blue.
Neutrophils and eosinophils are phagocytic, meaning they can engulf and digest pathogens,
whereas basophils are not. Neutrophils are the most abundant, making up 50-70% of circulating
leukocytes, while eosinophils make up 1-3%, and basophils and mast cells each make up less than 1%.
Neutrophils are produced in the bone marrow through hematopoiesis. Once released into the
bloodstream, they circulate for 7–10 hours before migrating into tissues, where they live for only a few
days. During infections, a large number of neutrophils are produced, and they are typically the first
cells to arrive at the inflammation site. This temporary increase in circulating neutrophils, known as
leukocytosis, is a medical indicator of infection. The process of neutrophils moving from the
bloodstream into tissues is called extravasation, which occurs in several steps:
★ Adherence to the vascular endothelium.
★ Penetration between endothelial cells lining the vessel wall.
★ Penetration through the vascular basement membrane into tissue spaces.
Neutrophil
Image source: Image source: Kuby Immunology 7th Edition
Eosinophils are mobile, phagocytic cells that can migrate from the bloodstream into tissue
spaces. While they have a phagocytic role in eliminating antigens, their significance in this function is
less than that of neutrophils. Eosinophils are involved in defending against multicellular parasitic
organisms, such as worms. The contents of their granules can damage parasite membranes, and they
often cluster around invading worms, releasing proteins that harm these parasites.
In addition to their role in parasitic defense, eosinophils, like neutrophils and basophils, can
secrete cytokines that regulate B and T lymphocytes, influencing the adaptive immune response. In
regions where parasitic infections are less common, eosinophils are more recognized for their
contributions to asthma and allergy symptoms.
Basophils are non-phagocytic granulocytes with large granules filled with basophilic proteins
that stain blue with standard H & E staining. They are naturally present in the body's circulation and
can be very potent. Basophils work by binding to circulating antibodies and releasing the contents of
their granules, which are pharmacologically active substances in their cytoplasm. These substances play
a key role in allergic responses. For example, histamines, which are common in basophilic granules,
increase blood vessel permeability and smooth muscle activity. Like eosinophils, basophils are
important in responding to parasites, particularly helminths (worms). Basophils also secrete cytokines
that help modulate the adaptive immune response.
Basophil
Image source: Kuby Immunology 7th Edition
Mast Cells
Mast cells originate in the bone marrow and are released into the bloodstream as
undifferentiated cells. They mature upon entering various tissues, where they can be found in the skin,
connective tissues of various organs, and mucosal epithelial tissues of the respiratory, genitourinary,
and digestive tracts. Like basophils, mast cells contain numerous cytoplasmic granules filled with
histamine and other pharmacologically active substances.
Mast cells play a crucial role in allergy development. Although basophils and mast cells share
many similarities, their exact relationship is not entirely understood. Some theories suggest that
basophils are the blood-borne equivalent of mast cells, while others propose that they have distinct
origins and functions.
Dendritic cell
Image source: Kuby Immunology 7th Edition
Dendritic cells perform two key functions: capturing antigens in one location and presenting
them in another. Immature dendritic cells patrol the body for pathogens outside the lymph nodes,
capture foreign antigens, process them, and then migrate to the lymph nodes to present the antigens to
naive T cells, initiating the adaptive immune response. As guards in peripheral tissues, immature
dendritic cells capture antigens through:
1) Phagocytosis
2) Receptor-mediated endocytosis
3) Pinocytosis
There are various types of dendritic cells, but their main function is to present antigens to T
helper (TH) cells. The four types of dendritic cells are:
a) Langerhans cells
b) Interstitial dendritic cells
c) Myeloid cells
d) Lymphoid dendritic cells
Each type originates from hematopoietic stem cells through different pathways and locations.
Despite their differences, they all express high levels of class II MHC molecules and co-stimulatory B7
family members. This makes them more potent antigen-presenting cells than macrophages and B cells,
which need activation to function as APCs.
Immature or precursor forms of these dendritic cells capture antigens via phagocytosis or
endocytosis. Once processed, the mature dendritic cells present the antigens to TH cells. During
microbial invasion or inflammation, both mature and immature Langerhans and interstitial dendritic
cells migrate to lymph nodes, where they present antigens to TH cells, initiating an immune response.
Follicular dendritic cells, which do not originate in the bone marrow, have a different role. They
do not express class II MHC molecules and therefore do not function as APCs for TH-cell activation.
Found exclusively in lymph node follicles, which are rich in B cells, these cells have high levels of
membrane receptors for antibodies. This allows them to bind antigen-antibody complexes, influencing
B cell responses.
Organs of the immune system
Introduction:
The immune system comprises a complex and essential network of various organs and
tissues, each with different functions in developing immune responses. These organs, known as
lymphoid organs, are involved in the growth, development, and deployment of lymphocytes.
Functionally, they are categorized into two main groups: primary and secondary lymphoid
organs. These organs are interconnected by the body's blood vessels and lymphatic systems,
creating a unified functional unit during immune responses against antigens by transporting
lymphocytes throughout the body, leading to systemic immunity. Additionally, tertiary lymphoid
tissues can import lymphoid cells during inflammatory responses.
1. Primary Lymphoid Organs:
These organs provide the environment for the development and maturation of lymphocytes
(e.g., white blood cells, leukocytes). The thymus and bone marrow are the primary (or central)
lymphoid organs. Immature lymphocytes generated during hematopoiesis mature and commit to
specific antigens in these primary organs. Mature lymphocytes residing in primary lymphoid
organs are immunocompetent, meaning they can mount an immune response.
Lymphoid organs
Image source: https://media.geeksforgeeks.org/wp-content/uploads/20240305100637/What-
Are-Lymphoid-Organs.png
1.1. Bone Marrow:
Bone marrow is the origin and development site for B-cells in most mammals, including
humans and mice. In birds, B-cell maturation occurs in the “bursa of Fabricius.” Mammals like
primates and rodents don't have a similar primary lymphoid organ, but in early gestation in cattle
and sheep, the fetal spleen hosts B-cell maturation, proliferation, and diversification. Later, this
function shifts to the ileal Peyer’s patch in the intestine, which contains a large number of B cells
(over 10 billion). Immature B cells proliferate and differentiate in the bone marrow after arising
from lymphoid progenitors. Stromal cells in the bone marrow interact directly with B cells and
secrete cytokines necessary for further development. During maturation, B cells with self-
reactive antibody receptors are eliminated through a selection process in the bone marrow.
Bone marrow
Image source: https://nci-media.cancer.gov/pdq/media/images/755927.jpg
1.2. Thymus:
The thymus is the first organ to produce lymphocytes during development, providing an
environment for T cell development and maturation. It's a flat, bilobed organ located above the
heart, with each lobe divided into lobules by connective tissue called trabeculae. Each lobule has
two compartments: the outer cortex, densely packed with immature T cells (thymocytes), and the
inner medulla, which has fewer thymocytes. Thymic lymphocytes are surrounded by epithelial
cells, dendritic cells, and macrophages.
In the outer cortex, some thymic epithelial cells, known as nurse cells, surround
thymocytes to form large complexes that influence their development. Dendritic cells and
macrophages are found in the medulla and at the junction between the cortex and medulla.
Epithelial cells in the thymus produce various cytokines needed for the differentiation of thymic
precursors into mature T cells. Thymocytes are attracted to the thymus from the bone marrow by
these cytokines. They mature in the cortex, migrate to the medulla, and are eventually released to
enter peripheral lymphoid tissues.
About 75% of all lymphocytes in the thymus are located in the deeper cortex.
Thymocytes in this area express CD1, CD4, and CD8 membrane molecules, unlike blood T cells,
which express either CD4 or CD8. Thymocytes undergo thymic selection, where T cells that
recognize self molecules are removed through apoptosis. Consequently, most T cells produced in
the thymus die. As they migrate to the medulla, they lose either CD4 or CD8 expression due to
genetic rearrangement. These naive, mature T cells then enter peripheral blood circulation and
are transported to secondary lymphoid organs, where they encounter and respond to foreign
antigens.
TS of Thymus
Image source: https://biologynotesonline.com/wp-content/uploads/2024/04/image-1479.png?ezimgfmt=ng:webp/ngcb1
2. Secondary Lymphoid Organs:
These organs capture antigens from specific tissues or vascular spaces and provide sites
for mature lymphocytes to interact with antigens effectively. The lymph nodes and spleen are the
primary secondary (or peripheral) lymphoid organs in the immune system.
2.1. Lymph Nodes:
Lymph nodes are encapsulated, bean-shaped structures that contain a reticular network of
lymphocytes, macrophages, and dendritic cells. Their main function is to filter antigens. Located
at the junctions of major lymphatic vessels, lymph nodes are the first organized structures where
immune responses to tissue antigens are initiated. Lymphatic vessels transport lymph to the
nodes, where antigens are filtered out. As the lymph is filtered in the nodes, it becomes enriched
with antibodies, cytokines, and mainly T lymphocytes.
Morphologically, a lymph node can be divided into three main concentric regions: the
cortex, paracortex, and medulla, each providing a distinct microenvironment.
a) Cortex: The outermost layer, mainly composed of B lymphocytes, macrophages, and
follicular dendritic cells arranged in clusters called primary follicles. Upon antigen
activation, these primary follicles enlarge into secondary follicles with a germinal center,
densely packed with proliferating B lymphocytes and plasma cells, along with
macrophages and dendritic cells. The germinal center is a crucial site for B-cell activation
and differentiation into plasma cells and memory cells.
b) Paracortex: Located beneath the cortex, this region is primarily populated with T
lymphocytes and some interdigitating dendritic cells. These dendritic cells express high
levels of class II MHC molecules, essential for presenting antigens to T helper (TH) cells.
c) Medulla: The innermost layer, sparsely populated with lymphoid-lineage cells, mainly
plasma cells actively secreting antibodies, along with activated TH and TC cells. This
region has a high concentration of immunoglobulins (Ig) due to the abundance of plasma
cells.
Structure of Lymph node
Image source: Janeway’s Immunology 9th edition
Lymph enters the lymph node through afferent lymphatic vessels, which pierce the node's
capsule at various points and empty into the subcapsular sinus. The lymph then flows slowly
through the cortex, paracortex, and medulla, allowing phagocytic cells and dendritic cells to
capture any bacteria or particulate material (e.g., antigen-antibody complexes) carried by the
lymph. The lymph exits the node through a single efferent lymphatic vessel, which is enriched
with antibodies and has a higher concentration of lymphocytes compared to the afferent vessels.
2.2. Spleen:
The spleen is a large, oval secondary lymphoid organ located high in the left abdominal
cavity. It filters blood and traps blood-borne antigens, responding to systemic infections. Blood-
borne antigens and lymphocytes enter the spleen through the splenic artery. The spleen is
encapsulated and extends projections called trabeculae, creating a compartmentalized structure
with two types of compartments: red pulp and white pulp, separated by a marginal zone.
Red Pulp: Contains red blood cells (erythrocytes) mixed with macrophages, dendritic
cells, a few lymphocytes, and plasma cells. This is where old and defective red blood
cells are destroyed and removed.
White Pulp: Surrounds the splenic artery, forming a periarteriolar lymphoid sheath
(PALS) mainly populated by T lymphocytes. The marginal zone, peripheral to the PALS,
is rich in lymphocytes and macrophages organized into primary lymphoid follicles.
The splenic artery carries blood-borne antigens and lymphocytes to the spleen, emptying
them into the marginal zone. Interdigitating dendritic cells trap the antigens and present them
with class II MHC molecules to T helper (TH) cells. The activated TH cells then further activate
B cells. Together with some TH cells, the activated B cells migrate to primary follicles in the
marginal zone and develop into secondary follicles with germinal centers, similar to those in
lymph nodes.
TS of Spleen
Image Source: https://biologynotesonline.com/wp-content/uploads/2024/04/image-
1483.png?ezimgfmt=ng:webp/ngcb1
3. Tertiary Lymphoid Tissues:
Tertiary lymphoid tissues generally contain fewer lymphoid cells than secondary
lymphoid organs. However, they play a crucial role in immune responses by rapidly increasing
lymphoid cell numbers during such responses. Key examples include mucosa-associated
lymphoid tissue (MALT), cutaneous-associated lymphoid tissue (CALT), and intraepithelial
lymphocytes (IEL).
3.1. Mucosal-Associated Lymphoid Tissue (MALT):
Mucous membranes lining the digestive, respiratory, and urogenital systems are
susceptible to pathogen attacks. These linings are protected by clusters of non-encapsulated,
organized lymphoid tissues known as MALT. These tissues range from loosely clustered
lymphoid cells in the lamina propria of intestinal villi to well-organized structures like the
tonsils, appendix, and Peyer's patches of the intestinal submucosa.
The lamina propria is rich in macrophages, B cells, plasma cells, and activated T helper
(TH) cells, providing stronger immunity compared to other body parts. MALT's critical role in
the body's defense is due to its large population of antibody-producing plasma cells. Notable
types of MALT include gut-associated lymphoid tissue (GALT) and bronchus-associated
lymphoid tissue (BALT), both of which are well-characterized.\
MALT
Image source: Kuby Immunology 7th Edition
3.1.1. Gut-Associated Lymphoid Tissue (GALT):
GALT, found in the gastrointestinal tract, is one of the best-characterized types of
MALT. It comprises Peyer's patches and isolated follicles in the tissue beneath the colon's
mucosa. Peyer's patches are lymphocyte aggregates forming primary lymphoid follicles (30-40
follicles with centrally located B cells surrounded by T cells and macrophages) that can develop
into secondary follicles with germinal centers upon antigen activation. These patches have
efferent lymphatics draining lymph into mesenteric lymph nodes but lack afferent lymphatics.
Peyer's patches are covered by specialized lympho-epithelium made up of microfold (M)
cells in small regions called inductive sites. M cells are flattened epithelial cells without the
microvilli typical of mucous epithelium, and they have deep pockets filled with clusters of B
cells, T cells, and macrophages. The epithelial cells of mucous membranes are crucial in
promoting the immune response by encountering antigens in the gut. M cells transport antigens
across the mucous membrane, activating B cells within lymphoid follicles. These activated B
cells then differentiate into IgA-secreting plasma cells. The antibodies are transported across
epithelial cells and released as secretory IgA into the lumen, where they interact with antigens
present. This is the site where secretory IgA is concentrated.
CALT
Image source: Kuby Immunology 7th edition
3.3. Intraepithelial Lymphocytes (IELs):
The outer mucosal epithelial layer contains intraepithelial lymphocytes (IELs), many of
which are T cells with unique receptors (γδT-cell receptors or γδTCRs) that have limited antigen
diversity and are well-positioned to encounter antigens entering through the intestinal mucous
epithelium. A large number of lymphocytes are associated with the epithelial surfaces of the
body, especially in the reproductive tract, lungs, and skin. The dermal layer of the skin contains
CD4+ and CD8+ T cells, as well as macrophages. Most of these dermal T cells are either
previously activated cells or memory cells. These collections of lymphoid cells play a crucial
role in developing mucosal immunity, contributing to both local and systemic immune responses
to antigens at the body surface.
Innate and adaptive immunity
The immune system is a complex network of immune cells and proteins like cytokines
working together to fight off pathogens such as bacteria, viruses, fungi, parasitic worms, and
abnormal cells. These cells have a remarkable ability to distinguish between self and non-self
molecules, although this ability can be compromised in conditions such as autoimmune diseases.
The differentiation between self and non-self is based on the Major Histocompatibility Complex
(MHC) proteins present on the surface of all body cells, except for identical twins.
There are two types of MHC proteins: MHC I and MHC II. MHC I is responsible for
differentiating the body’s own cells from foreign cells or pathogens and displaying antigens on the
cell surface. MHC II, found in antigen-presenting cells (APCs) along with MHC I, displays
phagocytosed microbes on the cell surface. MHC proteins are absent in red blood cells.
The body has two types of responses to invaders or antigens: Innate (Natural/Non-specific) and
Acquired (Adaptive/Specific).
1. Innate Responses:
These occur to the same extent regardless of how many times the pathogen is
exposed.
They employ phagocytic cells such as neutrophils, monocytes, macrophages, and
Natural Killer (NK) cells.
They respond immediately to foreign attacks.
They fight against all invaders, hence termed non-specific.
They comprise the first and second lines of defense.
2. Acquired Responses:
These responses improve with repeated subsequent exposures to foreign particles.
They involve antigen-specific B and T cells or antigen-presenting cells (APCs).
They take a longer time to react compared to innate responses.
They fight against specific types of invaders.
They include the third line of defense.
General principles and mechanism of innate immunity
Image
address:https://www.researchgate.net/publication/372822662/figure/fig1/AS:11431281180982350@
169770300515/General-principles-of-the-innate-immune-response-The-innate-immune-system-is-
a.png
Chemical Defenses
Chemical defenses in the body include chemicals and enzymes in body fluids, plasma
protein mediators, cytokines, antimicrobial peptides, and inflammation-eliciting mediators. These
elements destroy pathogens on the outer body surface, at body openings, and along inner body
linings.
1. Body Fluids:
Sweat, Tears, Mucus, and Saliva: Contain enzymes like lysozyme that break down
bacterial cell walls, killing the bacteria. Secretory IgA similarly attacks bacterial cell
walls.
2. Antimicrobial Peptides (AMPs):
Include dermcidin, cathelicidin, defensins, histatins, and bacteriocins. These AMPs
are produced in response to pathogens on the skin and help destroy them.
3. Cerumen (Ear Wax):
Contains fatty acids that lower the pH to between 3 and 5, protecting the auditory
canal from foreign particles like microbes.
4. Gastric Juice:
With a highly acidic pH of 2-3, it destroys pathogens that enter the stomach through
the oral cavity or nasal tract.
5. Urine Flow:
Acidic in pH, it helps kill microbes and flushes them out of the urethra.
6. Serum:
Contains unsaturated fatty acids that reduce water loss and inhibit microbial growth.
However, it also contains certain compounds that provide nutrition for some
microbes.
These chemical defenses work together to protect the body from a wide range of pathogens,
ensuring a robust defense against infections.
Chemical defenses of innate immune system
Image source: https://www.pinterest.com/pin/immunology-notes--70016969196713655/
Biological Defenses
Biological defenses are provided by living microorganisms that are friendly and beneficial.
These are known as resident natural flora and are found on our skin, in our bowel, and in other
places such as the mouth, gut, and reproductive parts. These microorganisms play a crucial role in
protecting our bodies from pathogenic invaders.
2. Complement Proteins:
a) These proteins circulate in the blood and, upon activation, enhance the ability of
antibodies and phagocytic cells to clear microbes and damaged cells from the
organism.
b) They promote inflammation and attack the pathogen's cell membrane, leading to
its destruction.
Key Mechanisms:
1. Phagocytosis:
Neutrophils, macrophages, and dendritic cells engulf and digest pathogens.
2. Natural Killer (NK) Cells:
These cells detect and induce apoptosis in virally infected or cancerous cells.
3. Inflammatory Response:
Immune and infected cells release cytokines that amplify cytokine secretion,
triggering an inflammatory response.
This response includes capillary dilation and increased permeability of the capillary
wall.
Macrophages help resolve the inflamed site by clearing up cellular debris.
4. Fever:
Cytokines increase core body temperature, resulting in fever.
This elevated temperature inhibits microbial growth and accelerates recovery and
repair processes.
5. Complement System:
Complement proteins in the blood serum are attracted to pathogens tagged by the
adaptive immune system.
A cascade of complement protein binding coats the pathogens, marking them for
detection by phagocytes and facilitating their digestion through phagocytosis.
These coordinated responses ensure that pathogens are effectively recognized and eliminated,
maintaining the body's defense against infections.
Lack of Immunological Memory:
Unlike the adaptive immune response, the second line of defense does not generate
immunological memory. This means it does not "remember" previous encounters with specific
pathogens, and the response is the same each time a pathogen is encountered.This second line of
defense is a critical part of our body's innate immune response, providing a rapid, non-specific
reaction to infections and helping to contain and eliminate pathogens before they can cause
significant harm.
Antigens are molecules or molecular structures that are foreign to the body and generally
induce an immune reaction by triggering the production of antibodies against them. As the
Antigens are substances that do not belong to the body, they are perceived as foreign invaders.
Antigens are typically defined by their ability to induce an immune response, but not all antigens
have the capacity to trigger such a response. The antigens that do induce a response are known as
immunogens.Antigens, indicated by the term 'Ag', can come in various forms including pollen,
viruses, chemicals, and bacteria.
The ability of antigens to elicit an immune response depends on specific regions called
antigenic determinants or epitopes. These determinants bind to receptor molecules with
complementary structures on immune cells to trigger a response.The concept of an antigen is
rooted in the body's ability to distinguish between its own components and foreign particles. This
distinction is crucial for an effective immune response.In response to antigens, the body produces
antibodies that specifically target and act against these foreign substances.Most antigens in
humans are proteins, peptides, or polysaccharides. However, lipids and nucleic acids can also
function as antigens when combined with proteins or polysaccharides.
Antigens might also be intentionally introduced into the body in the form of vaccines to
stimulate the adaptive immune system. This controlled exposure helps the body develop
immunity against specific pathogens.The presence of antigenic determinants on antigens allows
them to bind to receptor molecules on immune cells, leading to the activation of the immune
response. This process ensures that the body can identify and combat a wide array of pathogens
effectively.These insights highlight the crucial role of antigens in the immune system,
underpinning the body's defense mechanisms against a variety of foreign invaders.
Structure of Antigen
The structure of an antigen is characterized by its ability to bind to the antigen-binding site
of an antibody. Antibodies can differentiate between different antigens based on the unique
molecular structures present on the surface of the antigen.The molecular structure of an antigen
is characterized by its ability to bind to the antigen-binding site of an antibody.Most antigens are
proteins or polysaccharides. These can include coats, capsules, flagella, toxins, and fimbriae of
bacteria, viruses, or other microorganisms. Besides, secretions and other chemicals of the same
nature can also act as antigens.Lipids and nucleic acids of these microorganisms are only
antigenic when these are combined with proteins or polysaccharides.
Ideal immunogens are neither too large nor too small, with a molecular weight range of 8-
10K Da. Very small molecules can be made immunogenic by attaching them to larger molecules.
Antigens can be proteins, polysaccharides, lipids, or nucleic acids. The presence of aromatic
radicals is thought to enhance a molecule's antigenicity. Peptide antigens can generate a strong
immune response if they contain a high number of hydrophilic and charged amino acids such as
lysine, glutamine, glutamic acid, asparagine, and aspartic acid. These antigens should be
structurally different from the host's biomolecules and must interact easily with immune cells to
trigger an immune response.
The structure of antigens might be different depending on the nature of the antigen, their
size, and immunogenicity.All immunogenic antigens have a specific structural component called
epitope or antigenic determinant.The number of epitopes differs in different antigens and
determines the number of antibodies a single antigen can bind to.The region on antibodies that
interacts with antigens is called a paratope. It has been established that the structure of epitope
and paratope can be defined with a lock and key metaphor as the structures are specific and fit
with one another.
Apart from the chemical form, the physical form of a molecule can also influence its
immunogenicity. Particulate matter has been found to provoke a stronger immune response
compared to its soluble form. Interestingly, the denatured form of a biomolecule can elicit a
greater immune response than its native structure. Inert substances, such as dust, which cannot be
degraded by immune cells, tend to cause allergic reactions rather than antibody production.
Some of the structural Components of Antigens are:
1. Proteins and Polysaccharides:
Most antigens are proteins or polysaccharides. These can include components
such as coats, capsules, flagella, toxins, and fimbriae of bacteria, viruses, or other
microorganisms.
Secretions and other chemicals of a similar nature can also act as antigens.
2. Lipids and Nucleic Acids:
While lipids and nucleic acids are not typically antigenic on their own, they can
become antigenic when combined with proteins or polysaccharides.
3. Epitopes (Antigenic Determinants):
All immunogenic antigens possess specific structural components known as
epitopes or antigenic determinants.
The number of epitopes on an antigen varies, and this determines the number of
antibodies that can bind to a single antigen.
Epitopes bind to receptor molecules with complementary structures on immune
cells to elicit an immune response.
4. Paratopes:
The region on antibodies that interacts with antigens is called a paratope.
The relationship between an epitope and a paratope can be likened to a lock and
key metaphor, where the structures are highly specific and fit together precisely.
Properties of Antigens:
Antigens possess various properties that determine their immunogenicity, making them
crucial for understanding the immune response against them. Since these properties influence
immunogenicity, they are essential for identifying an effective antigen. Some of the key
properties of antigens include:
1. Foreign Nature
Antigens that trigger an immune response in the host are foreign to the recipient's
body.
The host body identifies the antigen as different from its normal components.
The immunogenicity of the antigen increases with the degree of foreignness. For
biological antigens, foreignness increases with the phylogenetic gap between
species.
However, exceptions exist, such as certain proteins within the host that may
induce an immune response, known as autoantigens.
Additionally, proteins and molecules from other species might not trigger an
immune response if they lack specific antigenic determinants or epitopes.
2. Chemical nature of antigens:
The most effective and commonly found antigens are proteins, followed by
polysaccharides.
Other molecules, such as lipids and nucleic acids, can also serve as antigens when
they form complexes with proteins and polysaccharides.
For proteins to act as antigens, they should have immunogenic regions containing at
least 30% of amino acids like lysine, glutamine, arginine, glutamic acid, asparagine,
and aspartic acid, along with a high number of hydrophilic or charged groups.
The level of immunogenicity increases with the heterogeneity of the molecules.
Homopolymers are generally less immunogenic than heteropolymers.
3. Molecular Size:
The molecular size of the antigens is also crucial in the immunogenicity of the
molecules.
It has been established that antigens should have a minimum size of greater than 5000
Da before they can be considered immunogenic.
However, low molecular weight substances can demonstrate immunogenicity when
coupled with large-sized carriers.
The low molecular weight substances are termed haptens that are considered ‘partial
antigens’ with at least one antigenic determinant.
4. Molecular Rigidity and Complexity:
The rigidity and complexity of molecules are essential factors that determine
immunogenicity.
In general, rigid molecules are good antigens as they can raise antibodies to certain
structures when compared to the less rigid ones.
The complexity of the structure is also an essential factor as a peptide antigen with a
repeating unit of a single amino acid is less immunogenic than a molecule with two or
more repeating amino acids units.
can lead to different responses to the same antigen in two strains of the same animal
species.
This variation arises from differences in the genes that govern the immune response,
such as the diversity in MHC alleles.
8. Degradability of antigens:
Antigens that are easily engulfed by phagocytes typically trigger a stronger immune
response.
Molecules with higher biodegradability are more effective as antigens. This is because
most antigens must be phagocytosed, processed, and presented to helper T cells by
antigen-presenting cells (APCs) to initiate an immune response.
Conversely, non-biodegradable substances like dust particles tend to cause allergic
reactions rather than an immunogenic response.
Types of Antigens:
Antigens can be categorized in various ways, with common classifications based on their origin
and immunogenicity.
1. Types of Antigens Based on Their Origin
Antigens can be classified into the following groups based on their origin:
a. Exogenous Antigens
Exogenous antigens are foreign substances that enter an organism's body from the
external environment. They can penetrate the body through ingestion, inhalation, or injection,
and are considered non-self-antigens. These antigens are found in body fluids and extracellular
spaces and are associated with MHC Class II molecules. Examples of exogenous antigens
include bacteria, viruses, allergens like pollen, and toxic foods.The uptake of exogenous antigens
is primarily mediated by phagocytosis via Antigen Processing Cells (APCs) like macrophages,
dendritic cells, etc. Some antigens initially enter as exogenous antigens but can become
endogenous, such as intracellular viruses.
Exogenous antigens
Image Source: https://ca.pinterest.com/pin/antigen-recognition-by-effector-t-cells--
419679259024475638/
b. Endogenous Antigens
Endogenous antigens are those that are generated within the cells of the host. These
antigens can be produced due to normal cell metabolism or as a result of infections by
intracellular pathogens such as viruses, bacteria, and parasites. Endogenous antigens may also be
the cells of the body or fragments, compounds, or antigenic products of metabolism.Endogenous
antigens might result in autoimmune diseases as the host immune system detects its own cells
and particles as immunogenic.
Endogenous antigens are processed by the macrophages and presented on the cell surface
by Major Histocompatibility Complex (MHC) class I molecules, which are then recognized by
cytotoxic T cells (CD8+ T cells).
Endogenous antigens
Image Source: https://ca.pinterest.com/pin/antigen-recognition-by-effector-t-cells--
419679259024475638/
c. Autoantigens
Autoantigens are proteins or protein complexes from the host that are mistakenly targeted
by the host's own immune system, leading to autoimmune diseases. This misdirected immune
response can be harmful, as the immune system attacks the body's own cells. The loss of
immunological tolerance to these antigens is influenced by genetic and environmental factors.
d. Tumor Antigens (Neoantigens)
Tumor antigens, or neoantigens, are displayed by Major Histocompatibility Complex
(MHC) I and II on the surfaces of tumor cells. These antigens arise from tumor-specific
mutations during the malignant transformation of normal cells. Typically, tumor cells develop
mechanisms to avoid antigen presentation and immune detection, preventing a robust immune
response.
e. Native Antigens
Native antigens are those that have not been processed by antigen-presenting cells
(APCs), which means that immune cells like T-cells cannot recognize them. However, B-cells
can be activated by these unprocessed antigens independently.
2. Types of Antigens Based on Immune Response:
Antigens can be categorized into two main groups based on their ability to elicit an immune
response:
a. Complete Antigens/Immunogens
Complete antigens, or immunogens, are capable of provoking a specific immune
response on their own. These antigens are typically proteins, peptides, or polysaccharides with a
high molecular weight, generally exceeding 10,000 Da.Some features of the complete antigens
are characteristics of complete antigens:
Self-Sufficient: These antigens can stimulate an immune response without the need for
polysaccharides.
Antigenic Determinants: Complete antigens have multiple antigenic determinant sites,
activate T cells, thereby playing a significant role in the body's defense mechanisms.
b. Incomplete Antigens/Haptens
Incomplete antigens, or haptens, are unable to generate an immune response by
themselves. They are usually non-protein substances that require a carrier molecule to form a
complete antigen. Haptens have a low molecular weight, generally less than 10,000 Da, and
possess fewer antigenic determinant sites. The carrier molecule attached to the hapten, usually a
protein or polysaccharide, is considered non-antigenic.
They are often non-protein substances, such as drugs, chemicals, or small organic
compounds.
Haptens possess fewer antigenic determinant sites compared to complete antigens.
The immune system recognizes the conjugate formed by the hapten and the carrier
molecule.
Once bound to a carrier, haptens can induce the production of specific antibodies
against the hapten.
Haptens are often used in research and vaccine development to create artificial
antigens.
Some drug allergies are caused by the immune system reacting to drug molecules
acting as haptens.
Haptens can form epitopes, the specific parts of an antigen recognized by antibodies
or T-cell receptors, when attached to a carrier.
The immune response is directed against both the hapten and the carrier molecule.
Conjugation to a carrier enhances the immunogenicity of haptens.
Common examples of haptens include penicillin, urushiol (the compound in poison
ivy), and certain cosmetic and food additives.
Haptens are valuable tools in immunological research for studying immune
responses.
Haptens are used in diagnostic tests to detect specific antibodies in the blood, such as
in allergy testing.
present them on their surface using molecules called MHC (Major Histocompatibility
Complex). T cells recognize these antigen-MHC complexes.
B Cell Receptors: B cells have membrane-bound antibodies (B cell receptors or BCRs)
that specifically recognize and bind to antigens. Each B cell has a unique BCR, tailored
to recognize a specific antigen.
2. Clonal Selection and Expansion
Activation: When a B cell's receptor binds to an antigen, the B cell is activated. Helper T
cells (specifically T_H cells) often assist in this activation by releasing cytokines.
Clonal Expansion: The activated B cell proliferates (clonal expansion) and differentiates
into plasma cells and memory B cells. Plasma cells produce large quantities of antibodies
specific to the antigen.
3. Antibody-Antigen Binding
Variable Region: The variable regions of the antibody (comprising the tips of the "Y"
shape) are highly specific to the antigen. They form a unique binding site that fits the
antigen's epitope (the specific part of the antigen recognized by the antibody).
Formation of Immune Complex: The binding of an antigen to an antibody forms an
antigen-antibody complex. This binding is highly specific, similar to a lock and key
mechanism.
4. Effector Functions of Antibodies
Neutralization: Antibodies can neutralize pathogens by binding to them and blocking
their ability to infect cells. For example, antibodies can prevent viruses from attaching to
host cells.
Opsonization: Antibodies coat the surface of pathogens, enhancing their recognition and
complement system. This leads to the formation of the membrane attack complex (MAC)
which can lyse (burst) the pathogen.
Antibody-Dependent Cellular Cytotoxicity (ADCC): Natural killer (NK) cells
recognize and bind to the Fc region of antibodies attached to infected cells or pathogens.
The NK cells then release cytotoxic molecules to kill the target.
5. Memory Formation
Memory B Cells: After the initial immune response, some B cells differentiate into
memory B cells. These cells persist in the body and can quickly result in a strong
response if the same antigen is encountered again in the future.
Understanding the complex structure of antigens and their interactions with antibodies is
essential for understanding how the immune system identifies and combats foreign invaders,
ensuring effective protection and immune surveillance.
Antigen – Antibody Interaction
Image Source: https://img.brainkart.com/imagebk39/8BsYwzt.jpg
B and T Cell Epitopes, Paratopes
B Cell Epitopes: Immunity Defenders
B cell epitopes, or antigenic determinants, are pivotal elements of the immune system.
They are specific regions of an antigen that are recognized by B cells, leading to an immune
response. The identification and characterization of B cell epitopes are critical for vaccine
development, diagnostics, and therapeutic interventions. B cell epitopes are part of the antigen
that binds to the B cell receptor (BCR) on the surface of B cells. This binding triggers a
cascade of events, leading to the activation, proliferation, and differentiation of B cells into
plasma cells that secrete antibodies. These antibodies specifically target and neutralize the
pathogen. The identification and characterization of B cell epitopes are critical for vaccine
development, diagnostics, and therapeutic interventions.
Epitopes:
Epitopes, also known as antigenic determinants, are specific parts of an antigen that
are recognized and bound by immune system components such as antibodies, B cell receptors
(BCRs), or T cell receptors (TCRs). They are critical for the immune system's ability to
identify and respond to pathogens.
1. Linear Epitopes
acids within the protein's primary structure. This sequence forms a specific linear
segment that antibodies recognize.
⬥ Recognition: These epitopes are recognized by antibodies based on their linear amino
acid sequence. The antibody's binding site, or paratope, interacts with this specific
sequence.
⬥ Stability: Linear epitopes remain recognizable to antibodies even when the protein is
⬥ Tertiary Structure: Conformational epitopes are composed of amino acids that are
not sequentially adjacent but come together in the protein's three-dimensional folded
structure. This spatial arrangement forms a unique shape recognized by antibodies.
⬥ Recognition: These epitopes are recognized by antibodies based on the protein's three-
dimensional structure. The antibody's paratope fits the specific shape of the
conformational epitope.
⬥ Stability: Conformational epitopes are dependent on the protein's native structure.
Denaturation or alteration of the protein's folding can disrupt the epitope, making it
unrecognizable to antibodies.
⬥ Example: In a folded protein, a conformational epitope might involve amino acids
from different parts of the primary sequence coming together in a specific spatial
arrangement
2. Antibody Production
Once activated, B cells differentiate into plasma cells that produce antibodies specific
to the recognized epitope. These antibodies circulate in the bloodstream and lymphatic
system, seeking out and neutralizing pathogens. The ability of B cells to produce antibodies
against specific epitopes is essential for the body to mount an effective immune response
against infections.
3. Immune Memory
B cell epitopes are also critical for the formation of immune memory. Memory B cells
are generated during the primary immune response and persist long-term in the body. Upon
re-exposure to the same antigen, these memory B cells rapidly respond by producing large
quantities of antibodies. This mechanism provides long-lasting protection against recurrent
infections and is the basis for the effectiveness of vaccines.
4. Vaccine Development
5. Diagnostic Applications
In diagnostics, B cell epitopes are utilized to detect the presence of specific antibodies
in a patient's blood. This can indicate exposure to a pathogen or the effectiveness of a
vaccination. Diagnostic assays, such as enzyme-linked immunosorbent assays (ELISAs),
often employ synthetic peptides representing B cell epitopes to capture and measure
antibodies.
6. Therapeutic Interventions
Monoclonal antibodies targeting specific B cell epitopes have been developed for
various therapeutic applications. These monoclonal antibodies can neutralize pathogens,
inhibit disease progression, and modulate immune responses. They are used in the treatment
of diseases such as cancer, autoimmune disorders, and infectious diseases.
Functions of B Cell Epitopes:
1. Recognition by B Cells
B cell epitopes are specific regions on antigens that are recognized and bound by B
cell receptors (BCRs) on the surface of B cells. This recognition is incredibly precise, as
BCRs have unique binding sites that match the epitopes. This specificity ensures that B cells
can accurately identify and bind to foreign pathogens.
The binding of an epitope to a BCR triggers a signaling cascade within the B cell,
leading to its activation. This process involves several steps:
Activation: The B cell receptor, upon binding to the epitope, sends signals to the B
3. Antibody Production
Plasma cells are the differentiated B cells that produce and secrete antibodies specific
to the epitope. These antibodies circulate throughout the body, binding to the same epitopes
on pathogens. This binding marks the pathogens for destruction by various immune
mechanisms. The specificity of antibodies ensures that they target only the harmful invaders
without affecting the body's own cells.
Role of b cell epitope in antibody production
Image source: https://webs.iiitd.edu.in/raghava/lbtope/images/home.jpg
During the immune response, some of the activated B cells differentiate into memory
B cells. These cells persist in the body long after the initial infection has been cleared. Upon
re-exposure to the same antigen, memory B cells quickly recognize and respond to the
specific epitopes, producing antibodies rapidly and in large quantities. This mechanism
provides long-term immunity and is the basis for the effectiveness of vaccines.
5. Enhancement of Phagocytosis
Antibody-bound epitopes can activate the complement system, a group of proteins that
work together to destroy pathogens. This activation triggers a series of reactions that lead to
the formation of the membrane attack complex (MAC). The MAC forms pores in the
membrane of the pathogen, leading to its lysis and destruction.
T cell epitopes are essential components of the immune system, playing a crucial role
in the activation and regulation of T cells.
1. Antigen Recognition
2. Activation of T Cells
The binding of a T cell epitope to a TCR triggers the activation of the T cell. This activation
involves several steps:
✿ Signal Transduction: The engagement of the TCR with the peptide-MHC complex
number of identical T cells (clones) that can recognize the same epitope.
✿ Differentiation: The expanded T cells differentiate into effector T cells (cytotoxic T
3. Effector Functions
✿ CD8+ Cytotoxic T Cells: These cells recognize epitopes presented by MHC class I
molecules. They secrete cytokines that help regulate the immune response by
enhancing the activity of other immune cells, such as B cells, macrophages, and
cytotoxic T cells.
4. Immune Memory
Some activated T cells differentiate into memory T cells, which persist in the body
long-term. These memory T cells provide rapid and robust responses upon re-exposure to the
same antigen. The presence of memory T cells is critical for long-lasting immunity and forms
the basis for the effectiveness of vaccines.
response to epitope recognition. These cytokines can enhance or suppress the activity
of other immune cells, ensuring a balanced immune response.
★ Immunoregulation: Regulatory T cells (a subset of CD4+ T cells) can recognize
6. Vaccine Development
The identification and utilization of T cell epitopes are essential for the development
of effective vaccines. Epitope-based vaccines aim to induce strong T cell responses by
including specific peptides that are recognized by T cells. This approach ensures targeted and
efficient immune protection against pathogens.
T-cell epitope mapping. (1) Immunogens are selected as screen targets; (2) T-cell epitopes are predicted
and tested using various bioinformatic methods; (3) T-cell epitopes are identified by various experimental
methods; (4) Acquisition of candidate epitopes.
Understanding T cell epitopes has led to the development of various therapeutic interventions:
that enhance the immune system's ability to target and destroy cancer cells.
⬥ Autoimmune Disease Treatment: Identifying epitopes involved in autoimmune
diseases can help develop therapies that modulate the immune response and reduce
tissue damage.
Paratopes:
Paratopes are the antigen-binding sites found on antibodies and T cell receptors
(TCRs). They are crucial for the immune system's ability to recognize and respond to specific
pathogens.
Paratopes are formed by the variable regions of the heavy and light chains of
antibodies or the alpha and beta chains of TCRs. These variable regions are the most diverse
parts of the molecule, generated through a process called V(D)J recombination.
★ Antibodies: Each antibody has two identical antigen-binding sites (paratopes) located
at the tips of the Y-shaped molecule.
a) The variable regions of both the heavy (VH) and light (VL) chains contribute
to the formation of the paratope.
b) The complementarity-determining regions (CDRs) within these variable
regions are particularly important for antigen binding. There are three CDRs in
each variable region, making a total of six CDRs per paratope.
a) The variable regions of the alpha (Vα) and beta (Vβ) chains form the paratope.
b) Similar to antibodies, the CDRs within the variable regions of TCRs are critical
for antigen binding, with three CDRs in each chain.
BCRs and antigens have direct interactions; interface residues are referred to as
"paratope" on the BCR side and "epitope" on the antigen side. On the other hand, TCRs
engage with peptide fragments originating from antigens, which are displayed by major
histocompatibility complex.
2. Diversity and Specificity
J segments.
★ Somatic Hypermutation (for antibodies): Introduces point mutations in the variable
regions of B cells during an immune response, increasing the diversity and affinity of
antibodies.
3. Antigen Binding
The paratope's structure is complementary to the epitope on the antigen, allowing for a
highly specific interaction. This binding is driven by various non-covalent forces, such as
hydrogen bonds, electrostatic interactions, Vander Waals forces, and hydrophobic
interactions.
a) Antibodies: The paratope binds to the epitope on the antigen, forming an antigen-
antibody complex. This binding can neutralize the pathogen or mark it for destruction
by other immune cells.
b) TCRs: The paratope binds to the peptide-MHC complex on the surface of antigen-
presenting cells. This interaction is essential for the activation of T cells and the
subsequent immune response.
4. Functional Roles
⬥ Antigen Recognition: Paratopes enable antibodies and TCRs to recognize and bind
where phagocytic cells (like macrophages and neutrophils) recognize and engulf the
antibody-coated pathogens.
⬥ Complement Activation: The antigen-antibody complex can activate the complement
malignant cells lead to the activation of cytotoxic T cells, which kill these target cells.
Functions of paratopes
Image source: https://ruo.mbl.co.jp/bio/e/support/method/images/antibody-sayou.png
5. Clinical Applications
specific antigens, using their paratopes to bind with high specificity. They are used in
treating cancers, autoimmune diseases, and infections.
⬥ Vaccines: Knowledge of paratopes and epitopes helps in designing vaccines that elicit
A hapten is an antigen but not an immunogen unless it is attached to a carrier protein. Immunogens are a
subset of antigens that can trigger an immune response on their own.
Image source: https://www.bosterbio.com/media/magefan_blog/Antibody_antigen_epitope_hapten_immunogen.jpg
Characteristics of Haptens:
Haptens are unique entities with specific characteristics that define their role in
immunology. These traits not only distinguish them from complete antigens but also explain
their behavior and utility in immune responses.
1. Size and Immunogenicity
Haptens are typically small molecules, often with molecular weights below 1,000
Daltons. Their small size and simple structure are insufficient to trigger an immune response on
their own. The immune system requires a certain degree of molecular complexity and size to
recognize a molecule as foreign and initiate an adaptive immune response.
However, when haptens are conjugated to a larger carrier protein, they form a composite
structure known as a hapten-carrier conjugate. This conjugate acts as a complete immunogen,
capable of activating immune cells. The carrier protein provides the necessary bulk and
complexity to ensure recognition by antigen-presenting cells (APCs) and subsequent T-cell
activation, while the hapten acts as a distinct epitope that is specifically targeted by antibodies.
For example, simple chemical groups like dinitrophenyl (DNP) or urushiol (found in
poison ivy) are not immunogenic in isolation. When these haptens bind to proteins in the body,
they become part of a complex that can elicit strong immune responses, often seen as allergic
reactions.
2. Binding Properties
Haptens are chemically reactive molecules capable of forming covalent bonds with
proteins. This property is critical for their role in immune activation. The conjugation process
involves the attachment of haptens to lysine residues or other functional groups on proteins,
forming stable hapten-carrier conjugates.
The covalent nature of the hapten-protein bond ensures that the hapten remains associated
with the carrier molecule during antigen processing. The conjugate is internalized by APCs,
where the protein component is broken down into peptides and presented on the cell surface in
association with major histocompatibility complex (MHC) molecules. Although the hapten itself
is not presented by MHC, its attachment to the carrier molecule ensures its presence in the
immune recognition process, leading to the generation of hapten-specific antibodies.
An example of this property is seen in drug-induced immune reactions. Penicillin, a small
molecule, binds covalently to serum proteins, forming hapten-carrier complexes that can provoke
immune responses.
3. Specificity
One of the most remarkable characteristics of haptens is their specificity. Once the
immune system generates antibodies against a hapten-carrier conjugate, these antibodies
recognize the hapten with high specificity, even in its free, unconjugated form. This reflects the
distinct chemical identity of the hapten, which acts as a unique antigenic determinant (epitope).
This specificity has significant implications in immunology and diagnostics. For instance:
In allergy testing, synthetic haptens identical to natural allergens are used to detect
specific antibodies in an individual’s serum.
In research, haptens like DNP or fluorescein are conjugated to antigens or proteins to
study immune responses or track biomolecular interactions using hapten-specific
antibodies.
⬥ Purpose of Conjugation: The carrier molecule provides the necessary size and
structural complexity that haptens lack, making the conjugate immunogenic. While
the carrier protein primarily facilitates immune system recognition, the hapten itself
acts as the specific epitope that will be targeted by antibodies.
⬥ Example:
In drug allergies, drugs such as penicillin act as haptens by binding to host proteins in
the bloodstream. The resultant hapten-carrier conjugate becomes the target of an
immune response, leading to hypersensitivity reactions.
2. Antigen Processing and Presentation
Once formed, the hapten-carrier conjugate is taken up by antigen-presenting cells
(APCs), such as macrophages, dendritic cells, or B cells, through a process called endocytosis.
✿ Internalization and Processing: After internalization, the carrier protein portion of the
of the APC, displaying the processed antigen to T-helper (CD4+) cells. It is important to
note that while the carrier protein is processed and presented, the hapten remains intact
and unprocessed. The presence of the hapten modifies the antigenic properties of the
carrier, effectively creating a unique antigen.
3. Activation of Lymphocytes
The interaction between the APC and T-helper cells marks a critical step in the immune
response. This stage involves two primary cell types:
★ T-helper Cell Activation: T-helper cells recognize the MHC class II-peptide complex
presented on the surface of the APC via their T-cell receptor (TCR). This recognition,
along with co-stimulatory signals provided by the APC, activates the T-helper cells. The
activated T-helper cells release cytokines that drive subsequent immune processes,
including B-cell activation.
★ B-cell Activation: B cells, which possess surface immunoglobulin (Ig) molecules as
antigen receptors, recognize and bind the hapten-carrier conjugate through the hapten
moiety. The internalized conjugate is processed by the B cell, and carrier-derived
peptides are presented on MHC class II molecules to T-helper cells. This interaction
provides additional activation signals to the B cell, leading to its proliferation and
differentiation into plasma cells.
★ Antibody Production: The plasma cells derived from activated B cells produce
antibodies specific to the hapten. These antibodies are highly specific and can recognize
and bind the hapten even in its free, unconjugated form. This specificity underlines the
immune system's ability to identify and remember molecular patterns.
★ Role in Drug Allergies: Small drug molecules like penicillin act as haptens by binding
act as haptens when they bind to skin proteins. The immune system recognizes these
hapten-protein complexes as antigens, resulting in delayed-type hypersensitivity reactions
and inflammatory responses, characteristic of contact dermatitis.
2. Vaccine Development
Haptens play a pivotal role in the design and development of conjugate vaccines,
especially for pathogens with antigens that are inherently non-immunogenic.
★ Polysaccharide Conjugates: Many bacteria, such as Haemophilus influenzae type b
antigens that act as haptens are conjugated to carriers to enhance immune recognition.
These therapeutic vaccines aim to stimulate the immune system to target and destroy
cancer cells.
Anti-tumor immunity based on the hapten theory of antibody production
Image source: https://www.creative-biolabs.com/native-antibody/static/img/1-1-1-3-Hapten-Carrier-Conjugation-1.jpg
3. Autoimmunity Research
Haptens serve as valuable tools in understanding autoimmune diseases, where the
immune system mistakenly attacks the body’s own tissues.
★ Hapten-like Behavior of Self-Molecules: In autoimmune diseases, small endogenous
molecules may act as haptens by binding to self-proteins and creating new antigenic
determinants. For example, in drug-induced lupus erythematosus, drugs such as
hydralazine can bind to DNA or nuclear proteins, forming immunogenic complexes that
trigger the production of autoantibodies.
★ Experimental Models: Researchers use haptens like trinitrophenyl (TNP) or
dinitrofluorobenzene (DNFB) to study immune responses in experimental models of
autoimmunity. These models provide insights into the mechanisms by which the immune
system distinguishes between self and non-self, as well as the conditions that lead to the
breakdown of self-tolerance.
Chemicals and drugs induce autoimmune-like responses via haptenation
Image source: https://www.mdpi.com/biology/biology-12-00123/article_deploy/html/images/biology-12-00123-g001.png
specific antibodies.
In allergy diagnostics, synthetic haptens identical to natural allergens are used to
For instance:
Haptenization of Tumor Cells: Tumor cells can be modified with haptens to
transplant rejection, haptens are being studied for their potential to induce
immune tolerance by modulating specific immune pathways.
⬥ Drug Design and Development: Understanding hapten-mediated immune reactions has
ADJUVANTS
Adjuvants are the enhancers of Immune Responses in Immunology. Adjuvants are non-
immunogenic substances that, when administered alongside antigens, significantly enhance the
immune system's response. The term "adjuvant" is derived from the Latin word adjuvare,
meaning "to help," aptly describing their function in immunological applications. These
compounds have transformed the fields of vaccine development and immunotherapy, improving
the efficacy of immunological interventions against infectious diseases, cancers, and
autoimmune disorders.
cytotoxic T cells (CD8+), which are critical for pathogen clearance and immune
regulation.
✿ Memory Cell Formation: By enhancing the activation and proliferation of
Adjuvants can boost vaccine efficacy in older adults, who are more susceptible to
infections.
✿ Infants and Immunocompromised Individuals: In these groups, where immune
Example Pathways:
a) TLR agonists (e.g., monophosphoryl lipid A) stimulate TLR4, leading to
the activation of nuclear factor kappa-light-chain-enhancer of activated B
cells (NF-κB).
b) This induces the production of pro-inflammatory cytokines (e.g.,
★ Promoted by adjuvants like TLR agonists, which stimulate IFN-γ and activate
★ Induced by adjuvants like alum, which promote IL-4 secretion and antibody
production.
Example: Alum-based vaccines for diphtheria and tetanus rely on Th2 responses
to neutralize toxins.
Mechanisms of action of adjuvants
Image source: https://media.springernature.com/lw685/springer-
static/image/art%3A10.1038%2Fnm.3409/MediaObjects/41591_2013_Article_BFnm3409_Fig4_HTML.jpg?as=webp
1. Inorganic Adjuvants
Inorganic adjuvants, composed mostly of salts and minerals, are commonly employed in
vaccines due to their ability to promote immune activation through depot formation or by
stimulating the innate immune system.
★ Aluminum Salts (Alum): The most frequently used adjuvants, aluminum salts like
aluminum hydroxide, aluminum phosphate, and aluminum sulfate, create a depot at the
injection site. This allows for slow release of the antigen, providing prolonged exposure
and enhancing immune activation. These adjuvants are particularly effective in triggering
Th2-type immune responses, resulting in high antibody production. Applications:
Diphtheria, tetanus, hepatitis B, and Hib vaccines.
★ Calcium Phosphate: Similar to aluminum salts, calcium phosphate forms a depot that
helps stimulate both innate and adaptive immune responses and is noted for improving
immune responses even with lower antigen doses. Applications: H1N1 influenza
vaccines and other pandemic flu vaccines.
3. Lipid-Based Adjuvants
These adjuvants are made from lipid particles that enhance immune cell activation,
particularly in stimulating cellular immunity.
★ ISCOMs (Immunostimulating Complexes): ISCOMs are lipid complexes formed from
cholesterol and other lipids, which encapsulate antigens and serve as a delivery system to
present the antigen to immune cells. They are highly effective in inducing both cellular
and humoral immune responses. Applications: HIV, malaria, and experimental cancer
vaccines.
★ QS-21 is a potent saponin-based adjuvant derived from the Chilean soapbark tree. It
activates both innate and adaptive immune responses, often inducing Th1 responses and
boosting antibody production. It is commonly combined with other adjuvants to enhance
their effectiveness. Applications: Cancer vaccines and malaria vaccines (under clinical
investigation).
4. Toll-Like Receptor (TLR) Agonists:
TLRs are critical components of the innate immune system, responsible for detecting
pathogen-associated molecular patterns (PAMPs). TLR agonists act as adjuvants by activating
TLRs and initiating immune cascades that enhance the immune response.
★ Monophosphoryl Lipid A (MPL): MPL is a derivative of lipopolysaccharide (LPS)
from bacteria, which stimulates TLR4 and activates a robust Th1 immune response,
enhancing cell-mediated immunity. Applications: Cervarix (HPV vaccine), malaria
vaccines, and cancer immunotherapies.
★ CpG Oligodeoxynucleotides (CpG ODN): CpG ODN are synthetic DNA sequences
that resemble bacterial DNA, recognized by TLR9. They stimulate a Th1 immune
response and promote cytokine production, such as interferon-gamma (IFN-γ).
Applications: Cancer vaccines and certain antiviral vaccines.
5. Saponin-Based Adjuvants:
Saponins are natural plant-derived compounds that can activate both humoral and cellular
immune responses. They are particularly effective in activating dendritic cells, which are crucial
for initiating immune reactions.
★ QS-21 (as mentioned earlier): QS-21 is a well-known saponin-based adjuvant that
boosts the immune system’s ability to recognize and process antigens, often used
alongside other adjuvants to enhance the overall immune response.
★ ISCOMs (Immunostimulating Complexes): ISCOMs include saponins along with
lipids, helping the immune system recognize and process antigens more effectively.
6. Bacterial-Derived Adjuvants
Some bacterial components also serve as adjuvants by activating immune responses and
bolstering the body’s defense mechanisms.
★ Bordetella pertussis toxin: This toxin, derived from the bacteria responsible for
T-cell activation, leading to stronger and more robust immunity. This is particularly
important for vaccines where both humoral (antibody-mediated) and cellular (T-cell-
mediated) immunity are needed.
✿ Reducing Antigen Dosage: By boosting immune responses, adjuvants allow for lower
doses of the antigen in vaccines. This can help stretch limited vaccine supplies, which is
especially useful during outbreaks or pandemics when rapid vaccine distribution is
necessary.
✿ Promoting Long-Term Immunity: Adjuvants enhance the generation of memory B-
populations.
c) Toll-Like Receptor (TLR) Agonists (e.g., MPL): Used in HPV and malaria vaccines.
adjuvants enhance the immune response against specific cancer antigens, improving
vaccine efficacy.
Examples in Cancer Immunotherapy:
a) ISCOMs (Immunostimulating Complexes): Used in experimental cancer vaccines.
b) QS-21: Combined with other adjuvants to enhance cancer vaccine responses.
The immunogenicity of vaccinations is improved by adjuvants. A T helper-polarizing cytokines, antibodies, and activated
T cells are produced in small amounts by vaccines devoid of adjuvants. B On the other hand, vaccines that contain
adjuvants encourage the maturation of more APCs, enhance the interaction between APCs and T cells, and encourage the
production of more T helper-polarizing cytokines, multifunctional T cells, and antibodies. These factors result in broad
and long-lasting immunity as well as dose and antigen savings. This figure was created with BioRender
(https://biorender.com/)
Image source: https://media.springernature.com/m685/springer-static/image/art%3A10.1038%2Fs41392-023-01557-
7/MediaObjects/41392_2023_1557_Fig1_HTML.png
3. Autoimmune Disease Research
Adjuvants are valuable tools in studying autoimmune diseases, providing insights into
how the immune system mistakenly attacks the body’s own tissues. By inducing inflammation
and immune activation, adjuvants help researchers explore the underlying mechanisms of
autoimmune responses.
✿ Autoantigen Presentation: Some adjuvants stimulate the immune system to produce
adjuvants provide long-term relief for individuals with allergic conditions like hay fever
or asthma.
Example:
a) Aluminum salts and oil-in-water emulsions: Used in vaccines for pollen or dust mite
allergies.
5. Infectious Disease Management
Adjuvants play an essential role in enhancing the immune system's ability to combat
infectious diseases. They are used to improve the efficacy of vaccines against a wide variety of
pathogens, including viruses and bacteria.
✿ Broadening Immune Responses: Adjuvants enhance both antibody production and
cellular immunity, enabling the immune system to target a broader range of pathogens
and improving vaccine efficacy.
✿ Improving Vaccine Efficacy in Special Populations: Adjuvants are particularly
important for vaccines targeting vulnerable groups, such as infants, the elderly, and
immunocompromised individuals, who may have weaker immune responses to vaccines.
Examples in Infectious Disease Vaccines:
a) MF59 and AS03: Used in influenza vaccines for at-risk populations.
b) MPL and CpG ODN: Used in vaccines for malaria, tuberculosis, and hepatitis.
viral infections, making them valuable in the development and treatment of HIV
vaccines.
✿ Autoimmune Disease Therapy: In autoimmune conditions, adjuvants can help adjust
immune responses to prevent the immune system from attacking healthy tissues.
Example:
a) ISCOMs and QS-21: Investigated in HIV and chronic viral infections.
7. Diagnostics
Adjuvants are used in diagnostic tools to improve the detection of specific antibodies or
antigens, enhancing the sensitivity of tests such as enzyme-linked immunosorbent assays
(ELISA) and lateral flow assays.
✿ Boosting Sensitivity: Adjuvants enhance the immune response to antigens or antibodies,
4 The stability and chemical composition: For recognition and processing, an antigen's
stability and integrity are essential. Long-lasting compounds that are highly stable can
interact with immune cells in an efficient manner. Antigens that are denatured or
degraded may no longer have the capacity to activate the immune system.
6 The dose of antigens: The immune response is affected by the quantity of antigen that is
injected into the body. An overly high dose can cause immunological tolerance or
suppression, while a dose that is too low may not be able to activate immune cells.
Frequently, there is a dose that maximises immunogenicity.
7 Administrative Route: The kind and intensity of the immune response are influenced by
the way an antigen is presented. As an example, Strong systemic reactions are often
elicited by subcutaneous and intramuscular methods. Weaker reactions or tolerance may
be brought on by intravenous methods. Localised immunological responses in mucosal
tissues can be triggered by mucosal channels, such as the nasal or oral.
receptors like Toll-like receptors (TLRs) and immunoglobulin genes can impact
recognition and response to antigens. For example, certain alleles may confer
susceptibility or resistance to infections or influence vaccine efficacy.
2. Age
Age significantly impacts the efficiency and nature of immune responses:
★ Neonates (Newborns): Neonatal immune systems are immature, with limited antigen-
Reduced production of naive T and B cells due to thymic involution and bone
marrow changes.
A shift towards a memory-biased immune repertoire, which may result in
cell function. These changes contribute to reduced vaccine efficacy and increased
susceptibility to infections.
3. Health Status
The overall health of an individual profoundly influences immunogenicity:
★ Immunocompromised States: Conditions such as HIV/AIDS, cancer, malnutrition, or
less robust immune response, characterized by the activation of naive T and B cells.
★ Secondary Exposure: Subsequent encounters with the same or cross-reactive antigens
trigger memory B and T cells, resulting in faster, stronger, and more specific immune
responses. This principle underlies the effectiveness of booster vaccines.
★ Cross-Reactivity: Previous exposure to structurally similar antigens can result in cross-
★ Sex Hormones:
Estrogens (predominantly in females) generally enhance immune responses,
contributing to the higher prevalence of autoimmune diseases in women.
Androgens (predominantly in males) tend to suppress immune responses, which
may contribute to differences in disease susceptibility and vaccine efficacy
between sexes.
★ Pregnancy: During pregnancy, the immune system adapts to tolerate the fetus (a semi-
allograft) by reducing T cell activity and pro-inflammatory responses. This can decrease
immunogenicity to certain infections or vaccines during this period.
6. Microbiome: The microbiome, the collection of microorganisms living in and on the human
body, profoundly influences immune system function:
★ Immune System Modulation: A diverse and balanced microbiome helps educate and
regulate the immune system, promoting effective responses to antigens while preventing
overreactions (e.g., allergies or autoimmune diseases).
★ Dysbiosis: Disruption of the microbiome balance (e.g., due to antibiotics, diet, or
★ Unhealthy Habits:
Smoking: Smoking damages epithelial barriers, increases oxidative stress,
and impairs the function of innate and adaptive immune cells, reducing overall
immunogenicity.
Alcohol Consumption: Excessive alcohol intake suppresses the immune
system by reducing cytokine production, impairing macrophage and T cell
functions, and depleting essential nutrients.
Physical Inactivity: Sedentary lifestyles are linked to inflammation and
reduced immune competence, while moderate regular exercise enhances
immunogenicity by improving circulation of immune cells and reducing
inflammation.
★ Environmental Pollutants: Exposure to air pollution, heavy metals, and industrial
chemicals can cause oxidative stress and inflammation, impairing immune responses
and reducing vaccine efficacy.
STRUCTURE OF ANTIBODIES
Antibodies, also called immunoglobulins, are protein molecules produced naturally by B-
lymphocytes. The term "antibody" refers to an immunoglobulin that specifically recognizes and
binds to an epitope on antigens. These soluble molecules, secreted by plasma cells, circulate
throughout the body to locate and attach to foreign substances, or antigens. When antibodies bind
to an antigen or microbial epitope, they can inhibit microbial activity and spread through various
mechanisms, such as immobilization, preventing microbial attachment to host cells, enhancing
phagocytosis, or marking microbes for destruction by soluble molecules or immune cells like
natural killer (NK) cells and eosinophils.
The primary role of antibodies is to recognize antigens and bind to them at their specific
antigen-binding sites. Once an antigen is detected, B-lymphocytes undergo proliferation and
differentiation into plasma cells, which then produce large quantities of antibodies to combat the
antigen. Their unique structure ensures antigen specificity, allowing them to bind exclusively to
their corresponding antigens. There are two classes of these antibodies:
1 Membrane bound antibodies: B-cell receptors (BCR) are composed of membrane-
bound antibodies, sometimes referred to as membrane-bound immunoglobulin (mIg) or
surface immunoglobulins (sIg). The surface of antibody-bound B-cells has receptors that
enable the B cell to recognise a particular antigen in the body. This activation causes the
B cell to multiply and develop into plasma cells, which generate the circulating
antibodies. The B-cell receptor (BCR) is typically made up of IgD or IgM antibodies that
are surface-bound and linked to Ig-α and Ig-β heterodimers, which can trigger signal
transduction. Between 50,000 and 100,000 antibodies are attached to the surface of a
single B-cell.
2 Freely circulating antibodies: The effector mechanisms of humoral immunity are
provided by freely circulating antibodies in the blood, which either hunt for and
neutralise antigens or mark them for removal by immune cells such as antigen-presenting
cells.
Structural Details:
Antibodies are large, globular, plasma proteins that weigh around 150 kDa and have a
diameter of 10 nm. Antibodies are glycoproteins because they are composed of glycans, which
are sugar chains that are attached to conserved amino acid residues. Glycans are essential for
maintaining the structure and functionality of antibodies. Additionally, they change the
antibody's affinity for the FcR (s). Immunoglobulin (Ig), the fundamental functional unit of an
antibody, is monomeric, whereas the released antibodies might be dimeric, tetrameric,
pentameric, or polymeric. The antibody is composed of constant C regions and variable V
regions.
Using myeloma proteins, Edelman for the first time extracted the antibody from a
multiple myeloma blood sample and initially identified its structure. The molecular weights of
the two chains they identified were 20 kDa for the light chain and 50 kDa for the heavy chain.
According to their respective concentrations, the fundamental antibody unit was made up of two
heavy and two light chains. Thus, the Antibodies are heterodimers. In IgG, IgD, and IgE, this
four-chain structure is present. In contrast, IgM exists as a pentamer with five basic units, and
IgA exists in both monomeric and polymeric forms (consisting of multiple basic four-chain unit
structures).
Immunoglobulin domains
The Immunoglobulin monomer has a “Y” – shaped molecule which has 4-polypeptide
chains; two identical heavy (H) chains and two identical light (L) chains which are
connected by a disulfide bond.
Each of the chains is made up of globular domains known as immunoglobulin domains
formed by intra-chain disulfide bonds. The domains containing 70-110 amino acids and
are classified into different types depending on size and function such as the variable
domain (IgV) and the constant domains (IgC).
The single variable region or domains (IgV) is of two forms, VL and VH consisting of 100
a sandwich shape, that is held together by the forces between the interaction of conserved
cysteines and other amino acids.
The domains have a particular immunoglobulin fold with two beta-sheets that form a
sandwich shape. The interaction of conserved cysteines and other amino acids holds the
sheets together.
General structure of Antibody
Image source: https://microbiologyinfo.com/wp-content/uploads/2015/05/Structure-of-Antibody2.jpg
By working in concert with the heavy chains, the light chains enhance the antibody's
ability to recognize and bind specific antigens, playing a critical role in immune defense. In
mammals, antibodies contain light chains that are of two types: lambda (λ) and kappa (κ). Each
light chain consists of two domains:
1. Variable (V) Domain: Responsible for antigen binding and varies among antibodies
produced by different B cells.
2. Constant (C) Domain: Conserved within a particular type of light chain (λ or κ) and
contributes to the structural stability of the antibody.
★ Each antibody molecule contains two identical light chains of the same type (either λ or
κ).
★ Only one type of light chain (either λ or κ) is present per antibody, regardless of the
heavy chain isotype. For example, an IgG antibody may pair its heavy chains with either
two λ chains or two κ chains but never a mix of both.
★ Light chains pair with heavy chains to form the antigen-binding sites of the antibody.
Together, the variable domains of the heavy and light chains create the specific structure
that recognizes and binds to antigens.
★ The light chain does not determine the antibody isotype but contributes to the diversity
★ Structurally, it includes:
One constant domain and one variable domain from both the heavy and light
chains.
This combination creates the antigen-binding sites that recognize and attach to
specific molecules.
★ The Fab region is essential for antigen recognition and specificity, as it ensures
antibodies can bind to their corresponding antigens with high precision.
2. Fv Region (Fragment Variable)
★ The Fv region is the smallest functional unit responsible for antigen binding.
★ It is located at the amino-terminal end of the antibody and includes the variable
domains of both the heavy (VH) and light (VL) chains.
★ The antigen-binding site, also called the paratope, is formed by this region. It interacts
directly with the epitope of the antigen.
★ Within the Fv region, the complementarity-determining regions (CDRs), which are
flexible loops, play a critical role in determining antigen-binding specificity.
CDRs are highly variable and allow the antibody to recognize a wide range of
antigens.
3. Fc Region (Fragment, Crystallizable)
★ The Fc region forms the tail of the antibody and consists of the constant heavy chain
domains (CH2 and CH3 in IgG, IgA, and IgE; CH2, CH3, and CH4 in IgM and IgE).
★ The Fc region is crucial for activating the immune system by interacting with:
Fc receptors on immune cells such as macrophages, natural killer (NK) cells, and
neutrophils.
Proteins of the complement system, initiating complement activation and
promoting immune responses such as opsonization and cell lysis.
★ Mediates effector functions like:
Phagocytosis: Enhancing the uptake of antibody-coated antigens by immune
cells.
Cytotoxicity: Directing cytotoxic immune cells to destroy pathogens or infected
cells.
Immune clearance: Facilitating the removal of immune complexes from the
bloodstream.
4. Complementary Roles
★ The Fab and Fv regions specialize in antigen recognition and binding, ensuring
antibodies can identify specific threats.
★ The Fc region bridges antigen recognition with immune system activation, enabling a
coordinated response to neutralize or eliminate the antigen.
★ Together, these regions allow antibodies to:
Bind specifically to antigens using the Fab region and paratope.
Activate immune mechanisms via the Fc region, triggering responses such as
inflammation, cell recruitment, and destruction of pathogens.
Immunoglobulin G – IgG
The most prevalent immunoglobin, IgG, makes up almost 80% of all serum antibodies.
IgG levels in the blood are roughly 10 mg/ml.
Structure
The basic structure of IgG is composed of a Y-shaped protein where the Fab arms are
linked to the Fc arms by an extended region of polypeptide chain called the hinge.The region is
exposed and sensitive to attack by proteases that cleave the molecule into distinct functional
units arranged in a four-chain structure.An IgG molecule consists of two identical γ heavy
chains, usually of the size 50kDa. The light chains in IgG exist in two forms; κ and λ, where the
κ form is more prevalent than λ, in the case of humans.
Subclasses of IgG:
Three subclasses of IgG antibodies have been identified: IgG1, IgG2, IgG3, and IgG4.
IgG1
It is the most prevalent of them, and they are called according to their serum quantity.
The most prevalent subclass of IgG antibodies with γ1 heavy chains is IgG1. Although soluble
protein antigen and membrane proteins are the main factors that produce IgG1, smaller amounts
of the other subclasses are frequently present as well. Since IgG1 is the most prevalent subtype, a
lack of it can result in a lower level of total IgG.
IgG2.
Composed of γ2 heavy chains, IgG2 is the second most prevalent kind of IgG in human
serum. Nearly all of the reaction to bacterial capsular polysaccharide antigens is mediated by
IgG2. The only subtype of IgG antibodies that is unable to pass through the placenta during
pregnancy is IgG2. IgG2 deficiency may lead to a weakened immune response to harmful
microbes.
IgG3
The third most prevalent kind of IgG seen in human serum is IgG3, which has γ3 heavy
chains. These are especially good at causing effector functions to be triggered. It is a short-lived,
strong pro-inflammatory antibody. With a strong affinity for FcR on phagocytic cells, IgG3 is
also the most efficient complement activator and facilitates opsonisation.
IgG4
With γ4 subclasses of heavy chains, IgG4 is the least prevalent IgG antibody in human
serum. IgG4 is produced in response to repeated or prolonged exposure to an antigen in a non-
infectious environment and is triggered by allergens. IgG4 has the ability to go from the mother
to the foetus across the placenta. Despite the rarity of IgG4 deficits, elevated serum IgG4 levels
have been linked to several issues in several organs.
Opsonisation
Image source: Abbas et al., Cellular and Molecular Immunology, 7e
3. Complement Activation
★ IgG can initiate the classical complement cascade by binding to antigens on the surface of
pathogens. When IgG binds to an antigen, it undergoes a conformational change that
allows it to interact with the first component of the complement system (C1q), leading to
the activation of the complement cascade.
★ Complement activation results in the formation of the membrane attack complex (MAC),
which can directly lyse pathogen cells, and the production of opsonins like C3b, further
enhancing phagocytosis.
4. Antibody-Dependent Cellular Cytotoxicity (ADCC)
★ IgG antibodies can bind to infected or tumor cells and recruit immune cells like natural
killer (NK) cells.
★ NK cells have Fc receptors that bind the Fc region of IgG, triggering the release of
cytotoxic molecules that kill the target cell. This is particularly important in fighting viral
infections and cancerous cells.
Antibody-Dependent Cellular Cytotoxicity (ADCC)
Image source: Abbas et al., Cellular and Molecular Immunology, 7e
5. Placental Transfer
★ IgG is the only antibody class that can cross the placenta, providing passive immunity to
the developing fetus. This transfer occurs through a receptor on placental cells, which
allows IgG to be transported across the placenta into the fetal bloodstream. This provides
the newborn with immediate protection against infections during the early months of life
until it’s immune system matures.
6. Regulation of Immune Responses
★ IgG antibodies also play a role in regulating the immune response. They can interact with
immune cells to fine-tune the production of other immune mediators. For instance,
certain IgG subclasses can influence T cell responses and modulate the activity of other
immune cells.
7. Memory Response in Adaptive Immunity
★ IgG is a major component of the immune memory generated after an infection or
vaccination. After initial exposure to a pathogen, B cells produce IgG antibodies, and
subsequent exposures result in a faster, more robust IgG-mediated immune response. IgG
is typically the predominant antibody produced during secondary immune responses,
which is key to long-lasting immunity.
Immunoglobulin M (IgM)
IgM is the third most abundant immunoglobulin in the serum, with a concentration of
approximately 1.5 mg/ml. It is the largest antibody and is the first to be produced during the
body's initial response to an antigen.
Structure of IgM
IgM is secreted primarily in a pentameric form, consisting of five subunits, each made up
of two µ heavy chains and two light chains. In some cases, a J chain may be present in the
hexameric form of the molecule, though it is not always included. The J chain is typically added
just before secretion, as it facilitates the polymerization of the individual monomers. Each
monomer contains two antigen-binding sites, giving the entire pentamer 10 binding sites.
However, not all of these binding sites can be occupied simultaneously due to spatial constraints.
The pentameric IgM has a molecular weight of 900 kDa.
Immunoglobulin A (IgA)
IgA, also known as sIgA (secretory IgA), is a crucial immunoglobulin primarily found in
mucosal areas of the body, where it functions as a protective antibody in various secretions.
Though present in relatively low concentrations in the blood, IgA is found in much higher
concentrations in bodily fluids like tears, saliva, sweat, and mucus. Its main role is to safeguard
mucosal surfaces from pathogens and foreign particles.
Structure of IgA
IgA has a molecular weight of around 160 kDa and is usually present as a monomer.
However, it can also exist in dimeric or trimeric forms, particularly in mucosal areas where it is
secreted to provide enhanced protection. The structure of IgA consists of four chains: two heavy
chains and two light chains.
Structure of dimeric IgA
Image Source: https://www.immunology.org/sites/default/files/inline-images/Immunoglobulin-A-IgA-Figure-1.png
✿ Heavy Chain Structure: The heavy chain of IgA is divided into three constant domains
(CH1, CH2, and CH3) and a variable region (VH) that is responsible for antigen
recognition.
✿ Hinge Region: The hinge region, located between the CH1 and CH2 domains, is a
flexible section that helps the antibody adapt its shape to better bind to antigens. This
region is held together by disulfide linkages, providing structural stability.
✿ Secretory Component: One of the defining features of sIgA is its secretory component,
which is an additional polypeptide chain of 75 kDa. This component helps in the
transport of IgA across epithelial cells and protects the antibody from degradation by
enzymes in bodily fluids, such as proteases.
✿ J Chain: IgA also contains a J-chain, which links the individual monomers in the dimeric
or trimeric form via disulfide bridges. The J-chain plays an important role in
polymerization and facilitates the secretion of IgA into mucosal surfaces, where it can
exert its protective function.
✿ IgA is divided into two subclasses: IgA1 and IgA2.
IgA1 exists primarily as a monomer and makes up about 80% of the IgA
found in serum. It is produced in the bone marrow and released onto mucosal
surfaces.
IgA2, on the other hand, is typically found in polymeric forms, including
dimeric structures, and is more commonly present in secretions produced
locally.
A key difference between IgA1 and IgA2 is the structure of their hinge
regions. In IgA1, the hinge region is notably longer and more extended
compared to IgA2.
Function of IgA
★ Mucosal Immunity: IgA is predominantly found in mucosal surfaces such as the
respiratory, gastrointestinal, and urogenital tracts, as well as in secretions like breast milk,
tears, and saliva. It serves as a first line of defense by preventing the attachment of
pathogens (like bacteria and viruses) to epithelial cells, thus blocking infections at these
entry points.
★ Barrier Protection: In its secretory form (sIgA), it forms a protective barrier against
harmful microorganisms and particulate antigens, neutralizing them and preventing their
penetration through mucosal membranes.
★ Protection from Enzymatic Degradation: The secretory component and the J-chain
confer protection to IgA against proteolytic enzymes that are abundant in mucosal
environments, ensuring that IgA maintains its functional integrity while performing its
immune functions.
Functions of IgA
Image source: https://labpedia.net/wp-content/uploads/2020/01/IgA-secretary-function1.jpg
Immunoglobulin D (IgD)
IgD is a monomeric antibody primarily found on the surface of immature B
lymphocytes, where it functions as a receptor for antigens. It is produced in small amounts and
secreted into the bloodstream, but its concentration in serum is much lower than other
immunoglobulins like IgG and IgM.
Structure of IgD
IgD exhibits a significant degree of structural diversity across vertebrate species,
adapting to fulfill its role alongside IgM in the immune system. Its flexibility is crucial for
complementing the functions of other antibodies, particularly IgM.
✿ Basic Structure: IgD is a glycoprotein made up of two identical δ (delta) heavy chains
and two identical light chains. These chains are linked by disulfide bonds, forming the
classic Y-shaped structure seen in immunoglobulins.
✿ Membrane Anchoring: The IgD that is found on the surface of B lymphocytes has
additional amino acid sequences at the C-terminal region, which anchor it to the cell
membrane. This membrane-bound form is essential for its role as an antigen receptor on
B cells.
✿ Disulfide Linkages and Domains: The light and heavy chains of IgD are connected by
interchain disulfide bonds. Additionally, there are intrachain disulfide bonds that
divide the chains into different structural domains. These domains contribute to the
stability and flexibility of the IgD molecule.
✿ Extended Hinge Region: One distinctive feature of IgD is its extended hinge region,
which provides increased flexibility to the antibody. This flexibility allows IgD to better
interact with antigens. However, the longer hinge region also makes IgD more
susceptible to proteolytic cleavage, which may limit its durability under certain
conditions.
2. Co-expression with IgM: On the surface of B cells, IgD is often co-expressed with
IgM, both of which serve as receptors for antigens. This co-expression ensures that B
cells have multiple ways of recognizing pathogens. IgM is more efficient at initiating
immune responses, but IgD plays a complementary role in fine-tuning the response.
Together, these antibodies collaborate to regulate the activation and differentiation of B
cells.
3. Regulation of B Cell Tolerance: IgD has been suggested to play a role in immune
tolerance, helping to prevent B cells from attacking the body's own tissues. This function
is essential for maintaining immune homeostasis and preventing autoimmune reactions. It
is thought that IgD may influence the selection and survival of B cells in the bone
marrow, ensuring that only B cells capable of recognizing foreign pathogens, rather than
self-antigens, are allowed to mature and enter circulation.
4. Activation of Signaling Pathways: When IgD binds to an antigen, it activates
intracellular signaling pathways in B cells, leading to changes in gene expression that
promote B cell activation, survival, and proliferation. This signaling is crucial for
initiating the adaptive immune response and ensuring that the immune system can mount
an appropriate response to infections.
5. Mucosal Immunity: While IgD is primarily found in the blood and on B cell surfaces, it
may also have a role in mucosal immunity, where it can participate in the local immune
responses within mucosal tissues, such as those in the respiratory and gastrointestinal
tracts. Some evidence suggests that IgD may help in the defense against pathogens by
interacting with mucosal epithelia, though its role in this context is less well understood.
6. Facilitation of B Cell Memory Formation: IgD is thought to play a role in the
development of B cell memory. After the initial antigen encounter, IgD may help in the
differentiation of B cells into long-lived memory cells. These memory B cells are crucial
for providing rapid and robust immune responses upon subsequent exposure to the same
pathogen.
Immunoglobulin E (IgE)
IgE (Immunoglobulin E) is a class of antibody primarily involved in the body’s immune
response to allergens and parasitic infections. It is present in very low concentrations in the
blood, but its functions are critical, especially in allergic reactions and defense against parasitic
organisms such as worms. IgE is best known for its role in hypersensitivity reactions, including
asthma, hay fever, and anaphylaxis. When an individual is exposed to an allergen, IgE binds to
the surface of mast cells and basophils through high-affinity receptors (FcεRI), leading to the
release of histamines and other inflammatory mediators upon subsequent exposures, to the same
allergen.
Structure of IgE
IgE, like other antibodies, has a basic Y-shaped structure consisting of two heavy
chains and two light chains. However, there are specific features that differentiate IgE from
other immunoglobulins, such as IgG, IgA, and IgM.
✿ Heavy Chains: IgE is composed of two heavy chains labeled ε (epsilon). These heavy
chains are longer than those found in other immunoglobulins, and they contain four
constant domains (CH1, CH2, CH3, and CH4). The CH3 and CH4 domains of the
heavy chain play key roles in binding to the high-affinity FcεRI receptors on mast cells
and basophils.
✿ Light Chains: Like other antibodies, IgE has two light chains, which can be of either
kappa (κ) or lambda (λ) type, and they are linked to the heavy chains by disulfide
bonds.
✿ Hinge Region: The hinge region in IgE provides flexibility to the molecule, allowing it
to interact with receptors and bind to antigens. This region is somewhat different from
that of IgG and IgA antibodies, contributing to the specific functional interactions of IgE.
✿ Fc Region: The Fc region (the tail of the Y-shaped antibody) is a crucial part of IgE's
structure. It interacts specifically with the FcεRI receptor found on mast cells and
basophils. This interaction is what enables IgE to trigger the release of histamine and
other inflammatory mediators when an allergen binds to the IgE on these cells.
✿ Antigen-Binding Site: The Fab region (the arms of the Y-shaped antibody) contains the
antigen-binding sites, where IgE can specifically bind to allergens or parasitic antigens.
The variable regions of the heavy and light chains are responsible for recognizing and
binding to these antigens, initiating the immune response.
Properties of IgE
IgE is an essential immunoglobulin in the immune system, primarily involved in allergic
reactions and defending against parasitic infections. Although it is present in very low
concentrations in the bloodstream, its effects are potent, especially in triggering inflammation
and immune responses to allergens.
1. IgE is found in extremely low concentrations in the blood, accounting for only about
0.05% of the total immunoglobulins in the serum. Despite this, IgE has significant
biological activity due to its role in hypersensitivity reactions and defense against
parasitic organisms.
2. One of the most distinctive features of IgE is its **high affinity for binding to the FcεRI
receptor on mast cells and basophils. When IgE binds to these cells, it "primes" them to
react more rapidly upon subsequent exposure to the same antigen (allergen). This
interaction is crucial for the immediate hypersensitivity response seen in allergic
reactions. Upon re-exposure to the allergen, IgE bound to mast cells or basophils triggers
these cells to release histamine, cytokines, and other inflammatory mediators that cause
the symptoms of an allergic reaction.
3. IgE is the central antibody in allergic responses. When an individual encounters an
allergen for the first time, IgE is produced and binds to the surfaces of mast cells and
basophils. In subsequent exposures, the allergen binds to the IgE, triggering
degranulation of these cells and the release of chemicals like histamine, which lead to
the symptoms of allergic reactions, such as swelling, itching, and difficulty breathing (in
severe cases, anaphylaxis).
4. IgE also plays a key role in defending against parasitic infections, particularly those
caused by helminths (worms). During a parasitic infection, IgE is produced and binds to
the surface of the parasite. This binding activates immune cells, including eosinophils,
which release toxic granules that help to destroy the parasite. IgE thus contributes to
immune defense against parasitic invaders by recruiting and activating effector cells that
can directly damage the pathogen.
5. IgE has a relatively short half-life in circulation compared to other immunoglobulins,
typically around 2 to 3 days. However, its binding to mast cells and basophils prolongs
its activity, as it remains on these cells until it is triggered by antigen exposure.
6. IgE is predominantly found on the surface of mast cells and basophils, rather than
circulating freely in the blood. This restricted distribution is a key factor in its role in
localized immune responses, especially in the tissues of the skin, lungs, and
gastrointestinal tract, where allergic reactions commonly occur.
7. IgE’s functions are distinct from those of other immunoglobulin classes like IgG, IgM,
and IgA. It is specifically adapted to promote allergic responses and parasitic
immunity, unlike other antibodies, which are involved in different immune functions like
bacterial opsonization (IgG), mucosal immunity (IgA), and complement activation (IgM).
8. Due to its central role in allergies, IgE is a target in immunotherapy for allergic
conditions. Anti-IgE therapies (e.g., omalizumab) are used to block IgE binding to
mast cells and basophils, thus preventing the release of histamines and other
inflammatory mediators. This approach helps in treating conditions like asthma and
chronic urticaria (hives).
Functions of IgE
Immunoglobulin E (IgE) is a specialized antibody that plays a crucial role in the body’s
response to allergens and parasitic infections. Though it is present in very low concentrations
in the bloodstream, its functional importance is significant, especially in mediating allergic
reactions and in immune responses against parasites.
1. Mediating Allergic Reactions
IgE is central to the immune response in allergic diseases. When a person with an
allergy encounters an allergen for the first time, the immune system produces IgE antibodies
specific to that allergen. This IgE binds to the surface of mast cells and basophils, which are
immune cells involved in inflammation.
Sensitization: The first exposure to an allergen leads to IgE production specific to that
allergen. This IgE binds to FcεRI receptors on the surfaces of mast cells and basophils,
"sensitizing" them.
Subsequent Exposure and Degranulation: Upon later exposure to the same allergen,
the allergen binds to the IgE on the mast cells or basophils. This triggers cell
degranulation, releasing histamines, cytokines, and other pro-inflammatory mediators.
Allergic Symptoms: The release of these chemicals leads to the classic symptoms of
allergic reactions such as itching, swelling, redness, rashes, and respiratory issues (like
sneezing, wheezing, or anaphylaxis). For example, in hay fever, asthma, and hives, IgE-
mediated responses contribute significantly to the severity of the symptoms.
Anaphylaxis: In severe cases, IgE activation can lead to anaphylaxis, a life-threatening
allergic reaction that requires immediate medical intervention. The widespread release of
inflammatory mediators can cause severe vasodilation, bronchoconstriction, and shock.
IgE Mediated allergic response
Image source: https://cdn.ps.emap.com/wp-content/uploads/sites/3/2020/12/Fig-1-IgE-mediated-allergic-response-1024x825.jpg
Functions of MAbs:
Monoclonal antibodies (mAbs) have a wide range of functions depending on how they
are designed and what they target. These functions are leveraged in diagnostics, therapeutics, and
research. Some of the important functions of monoclonal antibodies:
1. Targeted Immune Response: One of the primary uses of mAbs is to enhance the immune
system’s ability to recognize and destroy cancer cells, pathogens, or other harmful entities.
★ Immune System Activation: mAbs can bind to cancer cells or infected cells and act as a
signal to the immune system, marking them for destruction. This process is called
antibody-dependent cellular cytotoxicity (ADCC), where immune cells such as natural
killer (NK) cells or macrophages recognize and destroy the antibody-coated cells.
★ Complement Activation: Some mAbs can activate the complement system, a part of the
immune system that enhances the ability of antibodies and phagocytic cells to clear
pathogens or damaged cells. This process is known as complement-dependent
cytotoxicity (CDC).
★ Checkpoint Inhibition: Certain mAbs target immune checkpoint proteins (like PD-1 or
CTLA-4) that normally inhibit immune responses. By blocking these checkpoints, mAbs
can help “unmask” cancer cells, allowing immune cells to attack them more effectively.
2. Direct Tumor Cell Killing: Some monoclonal antibodies are designed to bind directly to
tumor cell markers and induce cell death.
★ Blocking Growth Signals: mAbs can block the binding of growth factors to their
receptors on cancer cells. This prevents the cancer cells from receiving the signals that
promote their growth and survival. For example, trastuzumab (Herceptin) targets the
HER2 receptor on breast cancer cells and inhibits their growth.
★ Inducing Apoptosis: Certain mAbs are designed to directly induce apoptosis
(programmed cell death) in cancer cells by binding to specific cell surface proteins. For
instance, rituximab targets the CD20 antigen on B-cells, leading to their destruction and
apoptosis.
3. Delivery of Cytotoxic Agents: Conjugated monoclonal antibodies are used to deliver
cytotoxic agents directly to cancer cells.
★ Radiolabeled mAbs: These antibodies are linked to radioactive isotopes, allowing them
deliver powerful cytotoxic agents directly to the cancer cells. This method, known as
antibody-drug conjugates (ADCs), minimizes the side effects of chemotherapy on healthy
tissues. An example is brentuximab vedotin (Adcetris), which targets CD30 on
lymphoma cells and delivers a toxic chemotherapy agent to the targeted cells.
4. Blocking or Inhibiting Pathogen Activity: mAbs are also used to target infectious agents
like viruses, bacteria, and fungi.
★ Virus Neutralization: mAbs can neutralize viruses by binding to their surface antigens
and preventing them from entering host cells. This is particularly useful in viral infections
such as HIV, Ebola, and COVID-19. For example, casirivimab and imdevimab are
mAbs used to treat COVID-19 by blocking the spike protein of the SARS-CoV-2 virus
from binding to the human ACE2 receptor.
★ Targeting Bacterial Toxins: Certain mAbs can neutralize bacterial toxins, preventing
them from causing damage to the host. For example, Bezlotoxumab is a mAb used to
treat infections caused by Clostridium difficile by neutralizing its toxins.
★ Blocking Autoimmune Targets: mAbs can target and neutralize specific immune
system components that are causing tissue damage. For example, adalimumab
(Humira) is used to treat rheumatoid arthritis by blocking tumor necrosis factor
(TNF), a molecule involved in inflammation.
★ Reducing Inflammation: mAbs that target immune cells or inflammatory cytokines
can reduce chronic inflammation in diseases like inflammatory bowel disease (IBD),
psoriasis, and lupus. For instance, infliximab (Remicade) targets TNF-alpha to help
reduce inflammation in diseases like Crohn’s disease and ulcerative colitis.
7. Gene Therapy and Cellular Therapy: mAbs are also involved in experimental
treatments, such as gene therapy and cellular therapy.
★ Gene Delivery: Some mAbs are engineered to deliver genetic material to specific cells,
modifying a patient’s own T-cells to express a receptor that recognizes a specific cancer
antigen. Monoclonal antibodies can be used to select or enhance the T-cells that are most
effective in targeting the cancer cells.
8. Bispecific Antibodies: These are engineered mAbs that can bind to two different antigens
simultaneously. Bispecific antibodies are useful in cancer immunotherapy, where they
can bind one arm to a cancer cell antigen and the other to a T-cell antigen, effectively
bringing immune cells directly to the cancer cells. This enhances the immune response
against tumors.
Applications of MAbs:
Monoclonal antibodies (mAbs) have a wide range of applications in medicine, research,
and diagnostics due to their ability to target specific molecules with high precision. These
applications can be broadly categorized into therapeutic, diagnostic, and research uses. Below is
a detailed note on the various applications of monoclonal antibodies (mAbs):
1. Therapeutic Applications of Monoclonal Antibodies
Monoclonal antibodies are extensively used in the treatment of several diseases,
particularly cancer, autoimmune diseases, infectious diseases, and others. The therapeutic
potential of mAbs has revolutionized medicine by providing highly specific treatment options
with fewer side effects.
A. Cancer Treatment (Oncology)
Monoclonal antibodies have proven to be effective in the treatment of various cancers by
targeting specific antigens on tumor cells.
⬥ Immune Modulation: mAbs can stimulate the immune system to recognize and destroy
cancer cells. For instance, rituximab (Rituxan) targets CD20 on B-cells and is used for
treating non-Hodgkin lymphoma and chronic lymphocytic leukemia (CLL).
⬥ Targeted Therapy: mAbs can bind to specific receptors or proteins on cancer cells,
inhibiting their growth or inducing cell death. For example, trastuzumab (Herceptin)
targets the HER2 receptor, which is overexpressed in some breast and gastric cancers,
and blocks signaling that promotes cancer cell growth.
⬥ Antibody-Drug Conjugates (ADCs): These are mAbs linked to chemotherapy drugs or
other cytotoxic agents, allowing the drug to be delivered directly to the cancer cell.
Brentuximab vedotin (Adcetris) targets the CD30 antigen on lymphoma cells and
delivers a cytotoxic drug.
⬥ Radiolabeled mAbs: mAbs can be conjugated with radioactive isotopes to deliver
(Keytruda), target immune checkpoint proteins like PD-1 or PD-L1, which are used by
tumors to escape immune surveillance. These mAbs enhance immune responses against
cancer.
B. Autoimmune Diseases
Monoclonal antibodies are also used to treat autoimmune diseases where the immune
system attacks the body’s own tissues.
⬥ TNF Inhibitors: mAbs like infliximab (Remicade), adalimumab (Humira), and
autoimmune conditions like rheumatoid arthritis and systemic lupus erythematosus (SLE)
to deplete autoreactive B-cells.
⬥ Interleukin Inhibitors: mAbs like ustekinumab (Stelara) target interleukins (IL-12 and
IL-23) and are used to treat inflammatory diseases like psoriasis and Crohn’s disease.
C. Infectious Diseases
Monoclonal antibodies have been developed for the treatment and prevention of various
infectious diseases.
⬥ Virus Neutralization: mAbs can neutralize viruses by binding to their surface antigens,
preventing them from entering host cells. For example, casirivimab and imdevimab are
mAbs used in the treatment of COVID-19, targeting the spike protein of SARS-CoV-2 to
prevent viral entry.
⬥ Bacterial Infections: mAbs are used to neutralize bacterial toxins, such as
bezlotoxumab (Zinplava), which targets Clostridium difficile toxins to prevent
recurrence of infection.
⬥ Ebola: ZMAP, a combination of three monoclonal antibodies, was used in the treatment
PCSK9 inhibitors used to lower cholesterol levels in patients at high risk for heart
disease.
2. Diagnostic Applications of Monoclonal Antibodies
Monoclonal antibodies are widely used in laboratory diagnostics for detecting and
quantifying specific molecules, including pathogens, proteins, or other biomarkers.
A. Immunoassays: mAbs are key components of diagnostic tests that detect specific proteins,
pathogens, or biomarkers in biological samples. Common techniques include:
★ Enzyme-Linked Immunosorbent Assay (ELISA): mAbs are used in ELISA tests to
target antigens.
★ Lateral Flow Assays: mAbs are used in home pregnancy tests and rapid tests for
T and B cells both use surface molecules to recognize antigens, but they do so in different
ways. B cells, with antibodies or B-cell receptors, can directly recognize antigens on their own. T
cells, however, can only recognize antigen pieces displayed on the surface of other cells. These
pieces are held by a protein called the major histocompatibility complex (MHC), which is
encoded by the MHC genes. After a cell breaks down an antigen, the pieces are displayed on the
cell surface by the MHC molecule. The MHC acts as a vessel for presenting antigen fragments,
so T cells can recognize them with their T-cell receptors. The MHC is named for its role in
determining whether a transplanted tissue will be accepted or rejected between individuals.
Major Histocompatibility Complex (MHC) was first discovered by the immunologist Peter Gorer
in the 1930s.
The Major Histocompatibility Complex (MHC) is a genomic region found in all
vertebrates, responsible for coding proteins crucial for immune recognition. In humans, the MHC
is a gene cluster located on chromosome 6, producing proteins known as Human Leukocyte
Antigens (HLA). These MHC proteins are essential cell surface molecules that allow the
acquired immune system to identify foreign substances, which is key in determining
histocompatibility. The primary role of MHC molecules is to bind peptide antigens and present
them on the cell surface for recognition by the appropriate T-cells. Among the numerous genes
in the human MHC, those encoding class I, class II, and class III MHC proteins are particularly
important.
There are two main types of MHC molecules: class I and class II. While they have
similar final structures, they differ in how they form these structures. Class I and class II also
vary in which cells express them and the type of antigens they present. Class I MHC molecules
are found on all nucleated cells and present antigens from inside the cell, like viral proteins, to
CD8+ T cells, which kill infected cells. Class II MHC molecules are mainly found on antigen-
presenting cells (APCs) and present antigens from outside the cell, like fungi or bacteria, to
CD4+ T cells. These activated CD4+ T cells help fight extracellular infections.
Class I and Class II MHC molecules are similar membrane proteins with related
structures and functions. Both types have been purified, and their 3D structures have been
determined through x-ray crystallography. These glycoproteins play a key role in presenting
antigens by holding peptide fragments in grooves and displaying them on the cell surface for
recognition by T cells through T-cell receptors. On the other hand, Class III MHC molecules are
unrelated to Class I and II in both structure and function, though many are involved in other
immune response processes.
Importance of MHC:
The Major Histocompatibility Complex (MHC) is a vital component of our immune
system, playing a key role in recognizing potential threats. When a foreign substance enters the
body, it must be eliminated. MHC ensures a clear distinction between the body’s essential
components and harmful invaders. MHC doesn’t act directly on foreign elements like antibodies
do. Instead, it recognizes these foreign substances and sends a signal to T lymphocytes, which
are responsible for destroying the invaders.
The primary role of MHC is to relay the correct message to the T cells, which then take
action. Depending on the situation, T cells may destroy the infected cells or eliminate just the
harmful element. Without MHC, the immune system would struggle to recognize and destroy
foreign elements, leading to serious health issues. Sometimes, certain substances that are
beneficial to the body could be mistakenly removed. In this way, MHC is crucial for signaling to
the body’s defense system, ensuring the destruction of harmful invaders while protecting
essential components.
derived from the degradation of intracellular proteins like viral or tumor proteins—on the
surface of cells. This allows CD8+ T cells (cytotoxic T lymphocytes) to detect and react
to cells that are infected or transformed.
★ Once a T-cell receptor (TCR) on a CD8+ T cell binds to the peptide displayed by an
MHC Class I molecule, the T cell becomes activated. This activation triggers the T cell to
carry out its immune functions, including inducing apoptosis (programmed cell death) in
infected or cancerous cells, ultimately limiting the spread of the pathogen or the growth
of abnormal cells.
2. Immune Surveillance and Response to Intracellular Pathogens:
★ MHC Class I molecules play an important role in the body’s ability to detect
enabling CD8+ T cells to identify and destroy cells that harbor intracellular infections,
such as viruses or intracellular bacteria.
3. Self vs. Non-Self Recognition:
★ One of the key functions of MHC Class I molecules is to differentiate between self cells
(healthy, non-infected cells) and non-self cells (infected or abnormal cells). This
distinction is vital to prevent the immune system from mistakenly attacking the body’s
own tissues, which could lead to autoimmune diseases.
★ By presenting foreign antigens (from viruses, bacteria, or tumors) on the cell surface,
MHC Class I molecules allow the immune system to target only abnormal or infected
cells, while avoiding healthy ones.
4. Activation of Cytotoxic T Lymphocytes (CTLs):
★ The principal function of MHC Class I molecules is to activate cytotoxic T lymphocytes
(CTLs). Upon recognizing the antigen presented by MHC Class I, CD8+ T cells become
activated, multiply, and proceed to destroy target cells.
★ Cytotoxic T cells are particularly crucial in controlling viral infections and in combating
cancer cells, as they can kill cells that harbor infections or are undergoing malignant
transformation.
5. Contributing to Cellular Immunity:
★ MHC Class I molecules are integral to cellular immunity, a branch of the immune
system where T cells eliminate infected or abnormal cells directly. This is in contrast to
humoral immunity, where B cells produce antibodies that target pathogens in body
fluids.
★ Cellular immunity, and MHC Class I molecules in particular, are especially important in
defending against intracellular pathogens like viruses and some bacteria, which reside
inside host cells and are not easily targeted by antibodies alone.
6. Role in Tissue and Organ Transplantation:
★ In the context of organ transplantation, MHC Class I molecules are essential for
prevents the immune system from attacking the body’s own healthy cells under normal
conditions. This regulation is crucial for avoiding the development of autoimmune
diseases, where the immune system wrongly targets and damages the body’s tissues.
Antigen presentation by MHC class I molecule
Image source: https://media.geeksforgeeks.org/wp-content/uploads/20221107172812/AntigenPresentation.png
engulf pathogens.
⬥ Antigen Processing: After the pathogen is engulfed, it is broken down in intracellular
class II molecules inside the vesicles. The MHC class II molecules have a peptide-
binding groove that can accommodate these peptides.
⬥ Surface Expression: The MHC class II-peptide complexes are transported to the cell
leading to its activation and differentiation into different subsets of T helper cells (e.g.,
Th1, Th2, Th17, Treg) based on the cytokine environment.
⬥ Co-stimulatory Signals: In addition to the peptide-MHC interaction, other co-
stimulatory signals (e.g., CD80/86 binding to CD28 on T cells) are needed for full
activation. This ensures that the T cell response is appropriate and regulated.
3. Immune Response Regulation
Once activated, CD4+ T helper cells assist other immune cells in responding to the
infection or threat. They coordinate various aspects of the immune response by releasing
cytokines and influencing the activity of:
⬥ B cells: Th cells help B cells produce antibodies in response to pathogens.
⬥ Cytotoxic T cells (CD8+ T cells): Th cells enhance the killing ability of cytotoxic T
Th1 responses for intracellular pathogens like viruses, and Th2 responses for extracellular
pathogens like parasites).
6. Antigen Specificity and Memory
⬥ MHC class II molecules help generate immunological memory by allowing CD4+ T
⬥ MHC class II molecules are involved in presenting the vaccine-derived antigens to CD4+
T cells, which then help generate both humoral (antibody-mediated) and cellular
immunity.
complement pathway. In this pathway, Factor B binds to C3b, and the complex activates
the alternative C3 convertase, helping to amplify the complement response and defend
against pathogens.
2. Tumor Necrosis Factors (TNFs)
★ TNF-α (Tumor Necrosis Factor-alpha): This cytokine is involved in systemic
inflammation and is part of the body's response to infection, trauma, and cancer. It plays
a central role in immune system regulation and inflammation by inducing fever,
apoptosis, and the activation of other immune cells.
★ TNF-β (Lymphotoxin): TNF-β, also known as lymphotoxin, is closely related to TNF-α
proteins from aggregating. They are induced in response to stress (e.g., heat, oxidative
stress, infection).
★ Heat shock proteins like HSP70 and others are crucial for stabilizing newly synthesized
Cytokines are a diverse family of small signaling proteins that play an essential role in
mediating an organism's response to injury, infection, and inflammation. These proteins function
as molecular messengers, transmitting signals between cells to regulate immune responses,
inflammation, and tissue repair processes. Cytokines are secreted in minute amounts by various
cell types, including immune cells such as macrophages, T cells, B cells, and mast cells, as well
as non-immune cells like epithelial and endothelial cells. Once secreted, cytokines exert their
effects by binding to specific receptors on target cells, initiating intracellular signaling cascades
that influence gene expression. This process may inhibit or enhance the activity of particular
genes, depending on the context, allowing cytokines to fine-tune the immune and inflammatory
responses.
Unlike endocrine hormones that typically act systemically throughout the body, most
cytokines operate in a localized manner near the cells that produce them. This ensures a targeted
and efficient response. For example, antigen-presenting cells (APCs) such as dendritic cells
release cytokines that drive T-cell activation. Upon encountering an antigen, APCs initiate a
rapid intracellular biochemical cascade, leading to the transcription of numerous genes, including
those encoding cytokines and their receptors. The activated T cells, in turn, release additional
cytokines to amplify and direct the immune response.
Most cytokines are secreted by immune system cells, while others, like type I
interferons and tumor necrosis factor-alpha (TNF-α), are produced by non-immune cells,
such as epithelial cells. These proteins play a pivotal role in regulating the development,
differentiation, and function of immune effector cells. Additionally, certain cytokines possess
direct effector functions, such as inhibiting viral replication (e.g., interferons) or inducing cell
death in tumor cells (e.g., TNF-α).
Cytokines are critical not only in regulating immune responses but also in maintaining
tissue homeostasis. Their dysregulation can lead to various pathological conditions, such as
chronic inflammation, autoimmune diseases, or cancer. For example, excessive cytokine
production may result in a "cytokine storm," a severe inflammatory state associated with
conditions like sepsis or severe viral infections. Conversely, inadequate cytokine production can
impair immunity, increasing vulnerability to infections.
The study and therapeutic targeting of cytokines have become central to modern
medicine. Cytokine-based therapies, such as the use of monoclonal antibodies to block pro-
inflammatory cytokines like TNF-α in rheumatoid arthritis or the administration of interferons in
viral infections and cancer, highlight their importance in clinical applications.
Properties of Cytokines:
Cytokines possess several distinct properties that enable them to regulate and coordinate
immune and inflammatory responses effectively. These properties define their roles as key
mediators of cellular communication in the immune system.
1. Low Molecular Weight: Cytokines are small proteins or glycoproteins with low molecular
weights, typically ranging between 8 and 30 kDa. This allows them to diffuse easily
through tissues and interact with receptors on target cells.
2. Specificity: Cytokines are highly specific in their action. They bind to specific receptors on
the surface of target cells, initiating intracellular signaling cascades. The presence of the
appropriate cytokine receptor on a cell determines whether the cell will respond to a
particular cytokine.
3. Pleiotropy: A single cytokine can exhibit multiple biological effects on different cell types.
For instance, interleukin-6 (IL-6) can promote inflammation, stimulate antibody
production, and support tissue regeneration, depending on the context and target cell.
4. Redundancy: Different cytokines may have overlapping functions, meaning several cytokines
can perform similar tasks. For example, IL-1, IL-6, and TNF-α all promote inflammation.
This redundancy ensures that critical immune functions can still occur if one cytokine is
absent or deficient.
5. Synergy: Cytokines often act in a synergistic manner, meaning that their combined effect is
greater than the sum of their individual effects. For instance, interferon-gamma (IFN-γ)
and TNF-α work together to enhance macrophage activation.
Image source: Kuby Immunology., 7th edition
6. Antagonism: Some cytokines can counteract or inhibit the effects of others. For example, IL-
10 and transforming growth factor-beta (TGF-β) are anti-inflammatory cytokines that
suppress the activity of pro-inflammatory cytokines like IL-1 and TNF-α. This
antagonistic behavior helps maintain immune balance and prevent excessive responses.
7. Localized Action: Most cytokines act in a localized or paracrine manner, affecting cells in
their immediate vicinity. However, in certain conditions, such as severe infections,
cytokines may act systemically, leading to widespread effects like fever or septic shock.
8. Transient Secretion: Cytokine production is highly regulated and transient. They are only
secreted in response to specific stimuli, such as infections, injury, or other immune
challenges, and their levels decrease once the stimulus is resolved. This ensures that
immune responses are appropriately timed and do not persist unnecessarily.
9. Cascade Effect: Cytokines often initiate cascades where one cytokine induces the production
of other cytokines. For example, TNF-α can stimulate the release of IL-1 and IL-6 to
amplify the immune response. This cascading property allows for rapid and coordinated
immune activation.
Image source: Kuby Immunology., 7th edition
10. Dual Nature: Cytokines can have both beneficial and harmful effects. For instance, they are
essential for fighting infections and healing wounds, but their overproduction can lead to
inflammatory diseases or cytokine storms, as observed in severe infections like COVID-19.
11. Receptor-Mediated Action: Cytokines function by binding to specific cell surface
receptors, which triggers intracellular signaling pathways such as the JAK-STAT
pathway. These pathways lead to changes in gene expression, influencing cellular
activities like proliferation, differentiation, or apoptosis.
12. Immunoregulatory Functions: Cytokines play a vital role in regulating immune responses.
They can stimulate immune activation (pro-inflammatory cytokines like IL-1, IL-6, and
TNF-α) or suppress immune activity (anti-inflammatory cytokines like IL-10 and TGF-β)
to maintain balance and prevent immune overreaction.
13. Short Half-Life: Cytokines are typically short-lived molecules, ensuring their effects are
limited to the time and place needed. This transient nature prevents prolonged or excessive
immune responses that could damage healthy tissues.
2. Monokines:
Produced by mononuclear phagocytic cells (e.g., macrophages), these cytokines regulate
inflammation and innate immune responses.
3. Interleukins (ILs):
Act as communicators between leukocytes, mediating a wide array of immune and
inflammatory functions. For instance, IL-1 promotes inflammation, while IL-10
suppresses it.
4. Chemokines:
Specialized cytokines that guide the migration of leukocytes to infection or injury sites,
facilitating the recruitment of immune cells during an immune response.
5. Interferons
Interferons (IFNs) are a distinct group of cytokines known for their critical role in
defending the body against viral infections, regulating immune responses, and modulating
cellular growth. They are low molecular weight proteins that are secreted primarily by cells in
response to pathogen invasion, particularly viruses, but also bacteria, parasites, and cancerous
cells. As signaling molecules, interferons act by transmitting biochemical signals between cells,
activating pathways that inhibit the replication of pathogens and orchestrate immune defense
mechanisms.
fibroblasts.
Type I interferons are primarily involved in antiviral defense. They trigger the
It acts on nearby cells and tissues by binding to specific receptors, TNF receptor
more effectively.
2. Apoptosis and Cytotoxicity:
★ TNF can induce programmed cell death (apoptosis) in infected or cancerous cells,
3. Immunomodulation:
★ TNF helps modulate adaptive immunity by enhancing antigen presentation and
activating T cells.
★ It influences the survival, proliferation, and differentiation of immune cells,
bacterial and viral pathogens. It stimulates antimicrobial activity and the release
of reactive oxygen species (ROS) by immune cells.
T cells can identify and react to protein antigens which is crucial in immune response
processes called antigen processing and presentation. Antigens undergo this process by breaking
down into peptides, attaching to Major Histocompatibility Complex (MHC) molecules, and then
being displayed on the cell membrane. The process of antigen processing starts when antigen-
presenting cells (APCs), including B cells, dendritic cells, and macrophages, internalise the
protein antigens. Antigen processing involves the enzymatic breakdown of the antigens into
smaller peptide fragments inside these cells.
A peptide-MHC complex is formed when the antigens are converted into peptides and
then bind to MHC molecules in the cytoplasm of the cell. The cytoplasm of the cell processes the
peptides produced from endogenous antigens in the case of MHC Class I molecules. Tumour,
viral, bacterial, and cellular proteins are examples of these endogenous antigens. These peptides
are bound by Class I MHC molecules, which then display them on the cell surface. This process
is known as cytosolic pathway.
However, peptides from external antigens that are absorbed by the cell through
phagocytosis or endocytosis are bound by MHC Class II molecules. Extracellular bacteria,
viruses, and other foreign particles are examples of these exogenous antigens. The endocytic
pathway processes the antigens and breaks them down into peptides. The resultant peptides
attach to Class II MHC molecules and travel to the cell membrane, where CD4+ T helper cells
can encounter and recognise them. This process is called as endocytic pathway.
MHC Class I and Class II molecules are essential for T cell activation because they
deliver antigen-derived peptides. The key to trigger an immune response to particular antigen is
the interaction between the peptide-MHC complex on antigen-presenting cells and the T cell
receptor on T lymphocytes. T lymphocyte activation and proliferation, as well as the
coordination of many immunological effector mechanisms, are the results of a series of immune
reactions set off by this recognition.
This mechanism is essential to immune surveillance, infection prevention, and the
removal of aberrant or contaminated cells. Target cells are typically cells that have contracted an
intracellular pathogen, such as a virus. Targets may also include changed self-cells, such as
cancerous cells, ageing body cells, or allogeneic cells from a graft. It is also a basic component
of the adaptive immune response.
mediated reaction.
Type IV hypersensitivity, characterized by delayed redness and inflammation, points toward a
T-cell-mediated response.
By narrowing down the root cause of symptoms, healthcare providers can offer faster and more
accurate diagnoses, reducing the risk of misdiagnosis and enabling effective management of conditions
ranging from food allergies to autoimmune disorders.
Tailored Treatment:
The classification system guides the development and administration of treatments that are
specific to the type of immune response involved as mentioned below
For Type I hypersensitivity, antihistamines, corticosteroids, or epinephrine are used to
understanding diseases like systemic lupus erythematosus (SLE) and type 1 diabetes.
Allergy vaccines: Research into Type I hypersensitivity has supported efforts to create
specifically target components of hypersensitivity reactions, like cytokine inhibitors for Type IV
responses.
Type I Hypersensitivity
presenting cells (APCs) like dendritic cells capture and process the allergen.
These cells present the allergen to naïve helper T-cells (Th2), prompting their activation.
Th2 cells release cytokines like interleukin-4 (IL-4) and interleukin-13 (IL-13), which
and itching.
Leukotrienes and Prostaglandins: Enhance inflammation, prolong smooth muscle
Second exposure
respiratory, and gastrointestinal tracts) where allergen exposure is likely. When IgE binds to
mast cells, it primes them for explosive degranulation upon allergen exposure.
✿ Basophils: These are circulating cells analogous to mast cells. They amplify the allergic
★ Epithelial Damage: Prolonged exposure can lead to destruction of epithelial cells, particularly
3. Immunotherapy:
Allergen Desensitization: Gradual exposure to increasing amounts of the allergen to
Type II Hypersensitivity
Type II Hypersensitivity
Image source: https://labpedia.net/wp-content/uploads/2020/04/Type-11-reaction-phagocytosisopsonization.jpg
Image source: https://labpedia.net/wp-content/uploads/2020/04/Type-11-reaction-ADCC.jpg
Examples:
Autoimmune Diseases
1. Autoimmune Hemolytic Anemia (AIHA):
IgG or IgM antibodies target antigens on red blood cell (RBC) membranes. This leads to
2. Goodpasture Syndrome:
IgG antibodies bind to the basement membrane in the glomeruli of the kidneys and
3. ABO incompatibility:
ABO incompatibility is a classic example of Type 2 hypersensitivity, which is an
antibody-mediated cytotoxic reaction. This occurs when a person receives a blood
transfusion with an incompatible ABO blood type, leading to the destruction of the
transfused red blood cells (RBCs) by the recipient's immune system.
The binding of IgM antibodies to the incompatible RBCs activates the complement
system.
This leads to the formation of the Membrane Attack Complex (MAC), which lyses the
transfused RBCs.
Complement activation releases inflammatory mediators like C3a and C5a, which
recruit immune cells and amplify the inflammatory response.
The destruction of RBCs results in the release of hemoglobin into the bloodstream,
which can lead to complications like jaundice or kidney damage.
Type III
hypersensitivity
Type IV Hypersensitivity:
Type IV hypersensitivity, also known as delayed-type hypersensitivity (DTH), is a
cell-mediated immune response that takes several hours to days to develop after exposure to
an antigen. Unlike Types I-III hypersensitivity, which are mediated by antibodies, Type IV
hypersensitivity is entirely dependent on T-cells and their interaction with antigens. The
delayed nature of this hypersensitivity is a hallmark feature, as the immune response peaks
between 24 to 72 hours after antigen exposure.
Type IV Hypersensitivity
Image source:
https://www.researchgate.net/publication/360322516/figure/fig1/AS:11431281139761969@1680886792751/Mechanism-
of-action-for-type-IV-hypersensitivity-reaction-T4HR-that-occurs-within-the.tif
Introduction to concepts of autoimmunity,
Primary and secondary Immunodeficiencies
All individuals have some level of immune recognition of their own tissues. In fact, for
T-cells to mature in the thymus, they must be positively selected based on their ability to
recognize the body’s own MHC molecules. Additionally, in most people, except those with
severe immune deficiencies self-reactive B-cells, T-cells that recognize self-peptides presented
by self-MHC, and autoantibodies (antibodies that react with the body’s own components) can be
found in the bloodstream. In people without autoimmune diseases, these autoantibodies are
mostly low-affinity IgM types, commonly produced by CD5+ B-1 cells as part of the body’s
natural antibody defense.
The term autoimmune disease is used when this self-reactivity leads to tissue damage or
illness. Some level of harmless autoimmunity may play a beneficial role in clearing out old or
damaged cells and molecules. Therefore, a mild degree of autoimmunity is considered normal
and usually doesn't cause disease. During lymphocyte development in the central lymphoid
organs, gene rearrangements naturally lead to the creation of some lymphocytes that can
recognize the body’s own antigens. Normally, these self-reactive cells are either eliminated or
kept under control through various regulatory mechanisms. These processes establish self-
tolerance, ensuring that the immune system does not target the body’s own tissues. Self-
tolerance relies on the coordinated function of multiple mechanisms that act at various stages and
locations during immune system development. The immune system uses different strategies to
prevent the activation of self-reactive lymphocytes and to avoid damage to the body's own
tissues. Autoimmune diseases occur only when immune tolerance fails and harmful self-reactive
lymphocytes are not properly controlled or eliminated.
The concept of autoimmunity was first introduced in the early 20th century by Paul
Ehrlich, who referred to it as "horror autotoxicus." Autoimmune responses are similar to normal
immune reactions against pathogens, but instead of targeting foreign invaders, they are triggered
by the body’s own antigens—called autoantigens. These responses lead to the production of self-
reactive immune cells and antibodies, known as autoantibodies, which attack the body’s own
tissues. When these self-directed responses become uncontrolled, they can result in a range of
chronic conditions known as autoimmune diseases. The breakdown can occur through several
mechanisms, which are grouped into central, peripheral, genetic, and environmental factors.
These diseases vary widely in terms of how severe they are, which tissues are affected, and the
immune mechanisms responsible for the resulting tissue damage.
Causes of Autoimmunity:
1. Failure of Central Tolerance
Central tolerance takes place during early lymphocyte development in the thymus for T-
cells and the bone marrow for B-cells. Its primary role is to eliminate strongly self-reactive
lymphocytes before they can enter the circulation. T-cells in the thymus undergo positive
selection (for recognizing self-MHC) and negative selection (elimination if they bind too
strongly to self-antigens). B-cells in the bone marrow that bind to self-antigens are either
eliminated by apoptosis (clonal deletion) or edited via receptor editing (a new light chain gene
rearrangement) or made anergic (unresponsive).
2. Failure of Peripheral Tolerance
Even after central tolerance, some autoreactive cells escape into the periphery. Peripheral
tolerance mechanisms which act as a second line of defense to prevent autoimmunity are:
a. Anergy (Functional Inactivation): Autoreactive lymphocytes that recognize self-antigen
without the necessary co-stimulatory signals (e.g., CD28 binding to B7) become anergic or
functionally inactive. In inflammation, antigen-presenting cells (APCs) may provide co-
stimulation to autoreactive cells, activating them.
b. Regulatory T-cell (Treg) Suppression: Tregs, expressing FOXP3, suppress immune
responses by releasing IL-10 and TGF-β (anti-inflammatory cytokines), direct cell–cell contact
inhibition, consumption of IL-2 to stop effector T-cells from receiving the growth signals.
Defects in Treg development/function lead to loss of immune suppression.
c. Activation-Induced Cell Death (AICD): Chronic activation of self-reactive T-cells induces
apoptosis via Fas-FasL interactions. Defects in Fas pathway prevent this cell death, allowing
autoreactive cells to persist.
3. Molecular Mimicry:
Some pathogens contain antigens structurally similar to self-antigens. Immune response
against the pathogen may accidentally cross-react with self-tissue due to similar epitopes. For
example: Streptococcal M protein resembles cardiac myosin. Antibodies formed against the M
protein attack heart tissue which leads to Rheumatic fever
4. Epitope Spreading:
Initial tissue damage or immune response exposes hidden or cryptic epitopes which were
not previously exposed to the immune system. This leads to a broader immune response against
multiple self-antigens in the same or nearby tissues. For example: In Systemic Lupus
Erythematosus (SLE), Initial response may target nuclear proteins, later it spread to DNA,
histones, etc.
5. Neoantigen Formation:
Self-molecules can be altered chemically or structurally by drugs, infections, oxidative
stress. These modifications generate neoantigens i.e., newly formed self-antigens that now
appear foreign to the immune system.
6. Genetic Predisposition:
Genetics play a major role in susceptibility to autoimmunity. Many autoimmune diseases
are associated with specific HLA alleles, especially MHC Class II molecules involved in antigen
presentation. Some genetic factors affects co-stimulation and Treg function, modify T-cell
receptor signaling, affect T-cell survival and expansion.
7. Environmental factors often serve as critical initiators or amplifiers of autoimmune
responses, particularly in individuals with an underlying genetic susceptibility. These external
influences can disrupt immune homeostasis, promote inflammation, or modify self-antigens in
ways that break tolerance and trigger autoimmunity.
Some of the important factors are:
★ Infections (Viral and Bacterial): Pathogens can provoke autoimmunity leading the
chemicals, may modify self-proteins to create neoantigens that the immune system no
longer recognizes as self.
★ Smoking: A well-established risk factor for diseases like rheumatoid arthritis and
individuals, can lead to keratinocyte apoptosis, releasing nuclear antigens and triggering
or worsening cutaneous lupus erythematosus and systemic lupus.
★ Diet and Gut Microbiota: The composition of the gut microbiome and dietary patterns
★ mediated by direct cellular damage in which the immune system targets and destroys
specific cells, leading to tissue damage or organ dysfunction. Some examples are
1. Hashimoto's Thyroiditis: An autoimmune disorder affecting the thyroid gland,
resulting in hypothyroidism. The immune system produces antibodies targeting
thyroglobulin and thyroid peroxidase, impairing hormone synthesis.
2. Autoimmune Anemia: Includes conditions like autoimmune hemolytic anemia,
where antibodies attack red blood cells. Leads to anemia with symptoms like
fatigue, pale skin, and shortness of breath.
3. Insulin-Dependent Diabetes Mellitus (Type 1 Diabetes): the immune system
destroys insulin-producing beta cells in the pancreas. Causes hyperglycemia and
reliance on insulin therapy for management.
★ mediated by Stimulating or Blocking Autoantibodies. These conditions arise when
autoantibodies alter the function of receptors or proteins. Some examples are:
1. Graves' Disease: Autoantibodies stimulate the thyroid-stimulating hormone
(TSH) receptor, leading to excessive thyroid hormone production
(hyperthyroidism).
2. Myasthenia Gravis: Autoantibodies block the acetylcholine receptor at the
neuromuscular junction, impairing communication between nerves and muscles.
Characterized by muscle weakness, particularly affecting the eyes, face, and
swallowing.
B. Systemic autoimmunity
It refers to autoimmune conditions in which the immune system targets ubiquitous or
widely distributed self-antigens. Unlike organ-specific autoimmunity, which is confined to a
single tissue, systemic autoimmune diseases affect multiple organs and tissues simultaneously or
sequentially. Pathogenesis of Systemic Autoimmunity involves:
IMMUNODEFICIENCY
The immune system is an incredibly adaptable defense mechanism that has evolved to
shield animals from harmful microorganisms and cancer. It can produce a vast array of cells and
molecules that can specifically identify and eliminate an almost endless variety of foreign
threats. These components function together in a highly complex and dynamic network, rivaling
the intricacy of the nervous system.
Immunodeficiency refers to a condition where the immune system is unable to
effectively combat infections. This state is also termed immunocompromised.
Immunodeficiency disorders weaken the immune system’s capacity to protect the body from
foreign or abnormal entities such as bacteria, viruses, fungi, or cancer cells. As a consequence,
individuals may become susceptible to uncommon bacterial, viral, or fungal infections, as well as
lymphomas or other types of cancer. Immunodeficiency reduces the immune system’s ability to
monitor and eliminate cancerous cells, a process known as cancer immunosurveillance.
✿ Combined B and T Cell Deficiency: Disorders involving both B and T cell impairments
include Severe Combined Immunodeficiency (SCID), Wiskott-Aldrich Syndrome, and
Major Histocompatibility Complex (MHC) deficiency, also known as Bare Lymphocyte
Syndrome.
✿ Interleukin-12 Receptor Deficiency: People with this deficiency are very prone to
widespread mycobacterial infections (like tuberculosis). This happens because their
immune cells lack the interleukin-12 (IL-12) receptor, which is needed to trigger a Th1
immune response, a key defense against these types of infections. Without this response,
the body can't fight off mycobacteria effectively.
T- Lymphocyte Deficiency:
Primary immunodeficiency disorders caused by T cell deficiency affect the body’s
ability to fight infections, especially those caused by viruses, fungi, and intracellular bacteria. T
cells play a crucial role in cell-mediated immunity and also help activate B cells to produce
antibodies. When T cells are defective or absent, the immune system becomes severely
compromised. Common T cell-related immunodeficiencies include:
1. Congenital Thymic Aplasia (DiGeorge Syndrome):
Also known as DiGeorge syndrome, this condition results from a developmental defect
in the thymus, the organ where T cells mature. Because the thymus is missing or
underdeveloped, affected individuals have very few or no functional T cells.
Common features include frequent viral and fungal infections due to poor T cell function,
Congenital heart defects, facial abnormalities, Low calcium levels (hypocalcemia) due to
underdeveloped parathyroid glands DiGeorge syndrome is often caused by a deletion on
chromosome 22 (22q11.2 deletion).
2. Chronic Mucocutaneous Candidiasis (CMC):
CMC is a condition marked by persistent and recurrent Candida (yeast) infections,
especially on the skin, nails, and mucous membranes (like the mouth and genitals). It occurs due
to a specific defect in T cell function that impairs the immune response to fungal infections.
Although other parts of the immune system may work normally, the failure of T cells to respond
properly to Candida allows the fungus to grow unchecked.
3. Hyper-IgM Syndrome (linked to T cell signaling defects):
While primarily known for its B cell antibody switching defect, Hyper-IgM syndrome
can also involve problems with T cell function, especially CD40 ligand (CD40L) deficiency on
T helper cells. CD40L is needed to help B cells switch from producing IgM to other antibody
types (like IgG, IgA, or IgE).Without this interaction, IgM levels are abnormally high, other
antibody levels (IgG, IgA, IgE) are very low, Patients have normal T and B cell numbers but
impaired communication between them. This leads to increased susceptibility to bacterial and
fungal infections and a higher risk of autoimmune issues.
Combined B and T Cell Deficiency
Combined immunodeficiencies are a group of primary immunodeficiency disorders
where both B cells (which produce antibodies) and T cells (which coordinate the immune
response and fight infected or abnormal cells) are either missing or not functioning properly.
These are often severe disorders that appear early in life and make affected individuals highly
vulnerable to a wide range of infections. Because both arms of the adaptive immune system are
impaired, patients are at high risk for recurrent bacterial, viral, fungal, and protozoal infections,
Poor response to vaccines, failure to thrive in infancy and increased risk of autoimmune diseases
and cancers. Common examples of combined B and T cell deficiencies include:
1. Severe Combined Immunodeficiency (SCID)
SCID is often called a "bubble boy disease" due to the need for extreme protection from
infections. It is one of the most serious forms of immunodeficiency, appearing in infancy.
Caused by various genetic mutations (X-linked SCID is the most common form), leads to almost
no functional T or B cells, infants appear normal at birth but soon develop severe, recurrent
infections (e.g., pneumonia, chronic diarrhea, thrush) and poor growth and development
2. Wiskott-Aldrich Syndrome (WAS)
WAS is a rare X-linked disorder affecting the immune system and blood cells. Caused
by mutations in the WAS gene, which affects cytoskeletal function in immune cells resulting in
combined immunodeficiency (low T and B cell function), eczema and thrombocytopenia (low
platelet count leading to bleeding)
3. Major Histocompatibility Complex (MHC) Class II Deficiency / Bare Lymphocyte
Syndrome
This rare disorder involves a lack of expression of MHC class II molecules on immune
cells, which is essential for proper T cell activation. Without MHC II, helper T cells (CD4+)
cannot be properly activated, as a result, B cells cannot produce antibodies
Phagocyte Deficiency
Phagocytes are a type of white blood cell that play a key role in the innate immune
system. They detect, engulf (eat), and destroy invading pathogens like bacteria and fungi. The
two main types of phagocytes are neutrophils and macrophages. When phagocytes are
deficient in number or don’t function properly, the body struggles to fight off infections
especially those caused by bacteria and fungi. This condition is known as phagocyte
deficiency, a type of primary immunodeficiency disorder (PID). People with phagocyte
deficiencies often suffer from frequent and severe bacterial infections (especially skin, lungs,
and gastrointestinal tract) delayed wound healing, abscess formation and fungal infections.
Common phagocyte-related immunodeficiencies include:
1. Chronic Granulomatous Disease (CGD): CGD is a genetic disorder where phagocytes
(mainly neutrophils) cannot produce reactive oxygen species (ROS) needed to kill ingested
microbes. As a result, the body tries to isolate infections by forming granulomas (clusters of
immune cells). Patients suffer from repeated skin, lung, liver, and bone infections caused by
bacteria and fungi. Inheritance can be X-linked (most common) or autosomal recessive.
2. Leukocyte Adhesion Deficiency (LAD): In LAD, white blood cells (especially
neutrophils) are unable to move from the bloodstream to the site of infection because they
cannot stick properly to blood vessel walls. This leads to severe bacterial infections, delayed
wound healing, and absence of pus formation despite serious infections.
3. Chediak-Higashi Syndrome: A rare inherited disorder where phagocytes have giant
granules that interfere with their ability to digest pathogens. It is associated with partial
albinism, neurological problems, and recurrent infections. Patients may develop a life-
threatening condition called the accelerated phase, similar to a lymphoma.
Complement Deficiency
The complement system is a group of over 30 proteins in the blood that work together as
part of the innate immune system. These proteins help destroy pathogens, enhance
inflammation, and assist antibodies and phagocytes in clearing microbes from the body. When
there is a deficiency or dysfunction in one or more complement proteins, the immune system
cannot respond properly to infections, especially those caused by bacteria. This condition is
known as complement deficiency, and it is a form of primary immunodeficiency disorder
(PID). Depending on which component is missing, the effects can vary. Complement
deficiencies are generally classified based on which part of the pathway is affected into the
classical pathway, alternative pathway, lectin pathway, or terminal pathway.
Secondary immunodeficiencies
They arise as a result of various diseases, medical conditions, or treatments that suppress
or impair the immune system. Unlike primary immunodeficiencies, which are inherited,
secondary forms are acquired and are far more common. They typically develop due to
underlying pathological processes that compromise immune function or from the use of
medications with immunosuppressive properties. Secondary immunodeficiencies may be
categorized as (a) B-cell deficiencies, (b) T-cell deficiencies, (c) complement deficiencies, and
(d) phagocytic deficiencies. The most prevalent and well-known example is Acquired
Immunodeficiency Syndrome (AIDS), which is caused by infection with the Human
Immunodeficiency Virus (HIV) and leads to a progressive decline in immune competence. In
addition to AIDS, secondary immunodeficiency can result from:
Immunosuppressive therapies, such as chemotherapy, corticosteroids, or NSAIDs
Psychological stress and depression
Severe burns
Radiation exposure
Neurodegenerative conditions like Alzheimer’s disease
Autoimmune and inflammatory diseases, including celiac disease and sarcoidosis
Hematologic disorders like lymphoproliferative disease, Waldenström’s
macroglobulinemia, multiple myeloma, aplastic anemia, and sickle cell disease
Nutritional deficiencies
Aging
Malignancies
Chronic illnesses such as diabetes mellitus
General introduction to vaccines, Types of
vaccines, Immunization programme
Vaccines are one of humanity’s most powerful tools against infectious diseases,
transforming global health and saving countless lives over the centuries. They have shaped the
way societies combat illness, reduce mortality, and improve public health. This essay provides a
detailed overview of vaccines, their types, mechanisms, significance, and future potential. At
their core, vaccines are biological preparations designed to provide immunity against specific
infectious diseases. They achieve this by training the immune system to recognize and combat
pathogens, such as bacteria and viruses. Unlike treatments that address symptoms after an
infection occurs, vaccines work proactively to prevent the disease from taking hold in the body.
The principle of vaccination is the stimulation of an immune response. Vaccines typically
introduce antigens harmless fragments or weakened forms of the pathogen into the body. This
stimulates the immune system to produce antibodies and memory cells, equipping it to recognize
and destroy the actual pathogen if it is encountered in the future. The process of immunization
prepares the immune system by exposing it to a simulated threat, the body is prepared to mount a
rapid and effective defense when faced with the real one. This preventive mechanism forms the
basis of individual immunity and contributes to the broader concept of herd immunity. Herd
immunity, achieved when a large proportion of a population is vaccinated, provides indirect
protection to those who cannot receive vaccines due to medical reasons.
Vaccines undergo rigorous testing in clinical trials before they are approved for public
use. These trials assess their safety and effectiveness, and post-approval surveillance ensures that
any rare adverse effects are monitored and addressed. Mild side effects, such as fever or soreness
at the injection site, are common and signify the immune system’s active response.
History of Vaccines
The history of vaccines is a testament to scientific ingenuity and dedication to public
health. The journey began with variolation in ancient China during the 10th century, where
powdered smallpox scabs were used to prevent the disease. This practice spread to Asia, Africa,
and later Europe, with Lady Mary Wortley Montagu introducing it to England in the 18th
century. The modern era of vaccination emerged in 1796, when Edward Jenner discovered that
cowpox exposure provided immunity against smallpox, coining the term "vaccine." Jenner's
work laid the foundation for immunology. The 19th century saw major advances, including
Louis Pasteur's development of rabies and anthrax vaccines and breakthroughs in bacteriology
that led to vaccines for cholera and typhoid fever.
The 20th century ushered in the "Golden Age of Vaccines," marked by the creation of
polio vaccines by Jonas Salk and Albert Sabin, the eradication of smallpox in 1980, and the
introduction of vaccines for diseases like measles, mumps, rubella, and hepatitis B. In the 21st
century, innovations like mRNA technology brought rapid solutions, as seen with COVID-19
vaccines. Global health initiatives continue to focus on eradicating diseases like polio and
combating outbreaks of Ebola and Zika, showcasing the ongoing impact of vaccines.
Principle of Vaccination
The principle of vaccination is rooted in harnessing the body's natural immune system to
provide protection against specific diseases. It is based on the concept of stimulating an immune
response without causing illness, thereby equipping the immune system to fight off the real
pathogen when encountered. Some key principles include:
1. Stimulating the Immune System
Vaccines introduce antigens either weakened, killed, or harmless fragments of
pathogensinto the body. These antigens act as foreign invaders, prompting the immune system to
recognize the pathogen as a threat. They generate targeted immune response, including the
production of antibodies and activation of immune cells. The antigens presented in a vaccine are
designed to mimic those found on the surface of the actual pathogen, ensuring that the immune
response will be effective if the pathogen is encountered.
2. Formation of Immunological Memory
One of the fundamental principles of vaccination is the creation of immunological
memory. During the immune response, specialized cells called memory B cells and memory T
cells are formed. These memory cells persist in the body for extended periods sometimes for a
lifetime and "remember" the specific antigens. If the individual is exposed to the actual pathogen
later, these memory cells enable the immune system to respond rapidly and efficiently, often
preventing the onset of illness. Immunological memory makes vaccines a powerful tool for long-
term disease prevention.
3. Types of Immune Responses
Vaccines can trigger various types of immune responses. Humoral Immunity involves
the production of antibodies by B cells. Antibodies neutralize pathogens or mark them for
destruction by other immune cells. Cell-Mediated Immunity involves T cells, which destroy
infected cells and regulate other immune responses. This is particularly important for
intracellular pathogens, like viruses. Effective vaccines often stimulate both types of immunity
for comprehensive protection.
4. Herd Immunity
Another key principle of vaccination is herd immunity, which protects populations by
reducing the spread of a pathogen. When a significant portion of a community is vaccinated, the
pathogen's ability to spread is curtailed. This protects individuals who cannot be vaccinated, such
as newborns or those with weakened immune systems. Herd immunity underscores the collective
responsibility of vaccination, highlighting its impact beyond the individual.
5. Safety and Controlled Exposure
Vaccines work by providing controlled exposure to antigens, avoiding the risks
associated with actual infection. This controlled exposure ensures that the immune response is
strong enough to build immunity. The risk of adverse effects is minimized through careful
vaccine design and rigorous testing.
6. Boosting Immunity Through Multiple Doses
For some vaccines, achieving full immunity requires multiple doses or booster shots.
Primary Series involves initial doses that help establish the immune response and create
memory cells. Boosters reinforce immunity by re-exposing the immune system to the antigen.
Boosters are particularly important for vaccines that provide immunity against pathogens with
rapidly mutating antigens or waning immune responses over time.
Types of Vaccines
Vaccines can be categorized based on their formulation into various types, including
whole organism vaccines (either attenuated or inactivated), vaccines derived from purified
macromolecules, recombinant vaccines, DNA-based vaccines, and multivalent subunit vaccines.
1. Live Attenuated Vaccines
Live attenuated vaccines utilize pathogens that have been weakened, so they cannot cause
disease in healthy individuals. These pathogens closely resemble the natural infection, allowing
the immune system to respond robustly and effectively. The attenuated pathogen replicates
minimally within the body, creating an immune response similar to that induced by natural
infection. This stimulates both humoral immunity (antibody production by B cells) and cell-
mediated immunity (activation of T cells).
Examples: Measles, Mumps, Rubella (MMR), Varicella Vaccine, Yellow Fever Vaccine
Advantages:
★ Produces a strong and long-lasting immune response due to its similarity to natural
infections.
★ Often requires fewer doses or boosters for effective immunity.
★ Provides broad immunity, sometimes offering protection against related strains.
Disadvantages:
Not suitable for people with weakened immune systems, as their bodies might struggle to
control even the weakened pathogen.
Requires strict storage conditions, such as refrigeration, which can limit accessibility in
resource-poor settings.
Rarely, the attenuated pathogen can revert to a more virulent form, although this is
exceedingly uncommon with modern vaccines.
2. Inactivated Vaccines
Inactivated vaccines consist of pathogens that have been killed or rendered inactive using
methods like heat or chemicals. These vaccines are incapable of causing disease. Since the
pathogen cannot replicate, the immune response is stimulated by its structural components, such
as proteins or carbohydrates.
Examples: Polio Vaccine (IPV), Hepatitis A Vaccine, Rabies Vaccine
Advantages:
★ Extremely safe for all individuals, including those with compromised immune systems.
★ Stable under normal storage conditions, making them easier to transport and distribute.
Disadvantages:
Induces a weaker immune response compared to live attenuated vaccines, often requiring
multiple doses or boosters to achieve long-lasting immunity.
Focuses mainly on humoral immunity, with limited activation of cell-mediated
immunity.
3. Subunit, Recombinant, and Conjugate Vaccines
These vaccines target specific parts of the pathogen, such as proteins, polysaccharides, or
surface antigens. They provide focused immunity without introducing the whole organism. By
isolating critical antigens, these vaccines stimulate the immune system to produce targeted
antibodies against the pathogen.
Examples: Hepatitis B Vaccine, Human Papillomavirus (HPV) Vaccine, Pneumococcal
Conjugate Vaccine
Advantages:
★ Lower risk of side effects because they exclude the pathogen’s unnecessary components.
★ Safe for use in immunocompromised individuals.
★ Highly effective for preventing diseases caused by bacterial toxins or surface antigens.
Disadvantages:
Generally requires multiple doses or booster shots for sustained immunity.
Limited in scope, as they only target specific parts of the pathogen, which may not
provide protection against all its strains.
4. Toxoid Vaccines
Toxoid vaccines specifically target diseases caused by bacterial toxins rather than the
bacteria themselves. These vaccines are developed by inactivating the toxins (toxoids) through
chemical or physical means, rendering them harmless while maintaining their ability to stimulate
an immune response. When certain bacteria cause disease, it is often due to the toxins they
release rather than the bacteria themselves. Toxoid vaccines expose the immune system to these
inactivated toxins, teaching it to neutralize them in the event of future exposure.
Examples: Tetanus Vaccine, Diphtheria Vaccine
Advantages
★ Highly effective against toxin-mediated diseases.
Disadvantages
Immunity may weaken over time, requiring booster shots to maintain long-term
protection.
Limited to combating diseases caused by toxins, not the bacteria themselves.
5. mRNA Vaccines
mRNA vaccines represent a groundbreaking advancement in immunology, using
synthetic messenger RNA to guide the body’s cells in producing a harmless antigen, typically a
protein from the pathogen. This antigen prompts the immune system to develop a defense
mechanism. These vaccines deliver mRNA into the body, which is absorbed by immune cells.
The cells use this mRNA as a template to produce a piece of the pathogen (e.g., the spike protein
of SARS-CoV-2). This triggers an immune response without exposing the individual to the
actual pathogen.
Examples: Pfizer-BioNTech COVID-19 Vaccine, Moderna COVID-19 Vaccine
Advantages
★ Highly efficient at inducing strong immune responses, including both humoral and cell-
mediated immunity.
★ Can be quickly adapted for emerging diseases or variants.
Disadvantages
Require ultra-cold storage to remain stable, posing challenges for distribution in
resource-limited areas.
Being a newer technology, there is limited long-term data on their duration of immunity
and potential effects.
6. Viral Vector Vaccines
These vaccines rely on a harmless carrier virus (vector) to deliver genetic material from
the target pathogen into the body. The host cells then produce the antigen, leading to an immune
response. The vector virus is engineered to contain genetic instructions for creating a key protein
from the pathogen. Once inside the body, the vector infects cells, causing them to produce this
protein and trigger immunity.
Examples: AstraZeneca COVID-19 Vaccine, Ebola Vaccine
Advantages
★ Mimic natural infections, eliciting a strong immune response.
Disadvantages
Limited usage and clinical approval compared to other vaccine types.
May elicit a weaker immune response, necessitating booster doses or other
enhancements.
8. Combination Vaccines
Combination vaccines merge antigens from multiple pathogens into one formulation,
reducing the number of injections required. By integrating multiple antigens in a single shot,
these vaccines simplify immunization schedules and enhance vaccination compliance, especially
in children.
Examples : DTaP Vaccine -Protects against diphtheria, tetanus, and pertussis.
Advantages
★ Reduces the burden of multiple injections, particularly for infants and children.
Disadvantages
The combination of antigens may lead to stronger immune reactions in some individuals.
Complex formulations can increase manufacturing challenges.
Emerging and Experimental Vaccine Technologies
Transplantation refers to the medical procedure in which cells, tissues, or organs are
surgically relocated from one part of the body to another, either within a single individual or
between a donor and a recipient. This method is a critical intervention employed when an organ
or tissue is no longer able to function adequately due to damage caused by disease, injury, or
failure. Through transplantation, a damaged organ system can be replaced with a healthy
counterpart from a donor, restoring essential functions and improving the recipient's quality of
life.
Organ transplantation is a complex and major surgical operation, generally considered a
last resort when all other medical treatments have proven insufficient. Due to its intricacies, it
needs meticulous planning and execution, as well as careful post-operative monitoring to ensure
long-term success.
The immune system plays an indispensable yet challenging role in the transplantation
process. While its primary function is to detect and eliminate harmful foreign invaders such as
bacteria and viruses, these same immune mechanisms can unintentionally become a significant
obstacle. The transplanted organ or tissue, referred to as the graft, is often recognized as a
foreign entity by the recipient's immune system. This triggers a defense mechanism known as
transplant rejection, in which the immune system attacks the graft, ultimately leading to its
destruction if left unmanaged.
The severity of the immune response largely depends on factors such as the type of graft
being used and the genetic differences between the donor and recipient. For example, grafts with
greater genetic disparities are more likely to provoke strong immune reactions, increasing the
risk of rejection. To mitigate this risk, donor and recipient matching is conducted prior to
transplantation. By carefully assessing immune compatibility, healthcare professionals aim to
minimize genetic discrepancies and reduce the likelihood of rejection.
Advancements in immunosuppressive medications further enhance the success rate of
transplantations by inhibiting the immune response and allowing the graft to integrate with the
recipient's body. But, these treatments necessitate a delicate balance to avoid complications, such
as increased vulnerability to infections. Transplantation represents a remarkable achievement in
modern medicine, offering patients with end-stage organ failure or irreversible tissue damage the
possibility of renewal and recovery. The surgical expertise and immunological considerations
emphasizes the complexity and significance of this lifesaving procedure.
Transplants or Grafts are categorized based on the source of the transplanted tissue or organ:
1. Allografts: These involve the transfer of tissue from one individual to another within the
same species. The individuals are genetically distinct, which may lead to immune
responses or rejection if precautions are not taken. Allografts are commonly used in
procedures like kidney transplants.
2. Isografts: It a specialized type of allograft which include transplanted tissues between
identical twins. Since monozygotic twins are genetically identical, the likelihood of
rejection is minimal, making these transplants highly successful.
3. Autografts: These are transplants where tissue is moved from one location on an
individual's body to another. For instance, skin grafts applied to burn wounds or the use
of tissue from one part of the body to reconstruct another part fall under this category.
Autografts eliminate the risk of immune rejection as the tissue comes from the same
individual.
4. Xenografts: This category involves the transplantation of tissue or organs from one
species to another, such as transferring tissues from animals to humans. While xenografts
have unique challenges, including immunological barriers, they are being explored in
medical research and applications like heart valve replacements derived from pigs.
Types of grafts
Image source:
https://as2.ftcdn.net/jpg/04/30/68/89/1000_F_430688960_tyuUF6kBTllfOeNNfCXeOrJEaQYrVnQE.jpg
B) B Cell Involvement
B cells are another key player in graft rejection, particularly in antibody-mediated
rejection (AMR). These cells become activated after recognizing donor antigens and interacting
with helper T cells. Once activated, B cells differentiate into plasma cells, which produce
antibodies specific to the donor antigens. These antibodies can bind to donor cells, target them
for destruction via mechanisms such as complement activation and also recruit additional
immune cells, such as natural killer (NK) cells, through a process known as antibody-
dependent cellular cytotoxicity (ADCC). The production of these donor-specific antibodies
(DSAs) can lead to vascular inflammation and damage, playing a central role in acute and
chronic graft rejection.
Effector Mechanisms
Effector mechanisms include the various ways in which immune cells and antibodies
mediate the destruction of the graft. These mechanisms include:
1. Direct Cell Lysis: Cytotoxic T cells induce apoptosis in donor cells by releasing
cytotoxic molecules such as perforin (which creates pores in the target cell membrane)
and granzymes (which trigger cell death).
2. Inflammation: Helper T cells and other immune cells release inflammatory cytokines
(e.g., TNF-α, IL-1), which recruit macrophages and neutrophils to the graft site. These
cells exacerbate tissue injury by producing reactive oxygen species (ROS) and proteolytic
enzymes.
3. Antibody-Mediated Damage: Antibodies bind to endothelial cells lining the graft’s
blood vessels, triggering the activation of the complement cascade. This process results in
the formation of the membrane attack complex (MAC), which disrupts cell membranes
and damages graft vasculature. The complement system also generates inflammatory
mediators (e.g., C3a, C5a) that further amplify the immune response.
4. Vascular Disruption: The immune response against the graft's blood vessels can lead to
thrombosis (clot formation) and loss of blood supply, causing ischemia and ultimately
graft failure.
3. Treatment of Rejection: When signs of acute or chronic rejection are detected, more
aggressive strategies are employed to counteract the immune response and restore graft
function. High doses of corticosteroids, monoclonal or polyclonal antibodies, and
adjustments to maintenance therapy regimens are used during this phase. The treatments
aim to target activated immune cells and suppress inflammation.
Important Immunosuppressors:
Immunosuppressive drugs are essential in managing graft rejection, as they inhibit or
modulate the recipient's immune system to ensure the survival and functionality of the
transplanted organ or tissue. Each class of immunosuppressors targets specific pathways in the
immune system, and they are carefully combined to achieve optimal therapeutic outcomes.
1 Calcineurin Inhibitors: Calcineurin inhibitors block the activity of the enzyme
calcineurin, which is crucial for activating T cells. Calcineurin normally facilitates the
production of interleukin-2 (IL-2), a cytokine that promotes T cell proliferation. By
inhibiting calcineurin, these drugs effectively suppress the expansion of activated T cells,
a key component in the immune response against the graft. These are foundational drugs
in maintenance therapy and are highly effective in preventing acute rejection by limiting
T cell-mediated immune responses. Long-term use can lead to nephrotoxicity (kidney
damage), hypertension, increased cholesterol levels, and a heightened risk of infections.
Patients require frequent monitoring of blood levels to optimize dosing and minimize
toxicity.
Examples: Cyclosporine, Tacrolimus.
2 Corticosteroids: Corticosteroids reduce inflammation by inhibiting the production of
pro-inflammatory cytokines such as TNF-α, IL-1, and IL-6. They also interfere with the
activation and functioning of immune cells, including T cells, B cells, and macrophages,
thereby dampening the immune response against the graft. Corticosteroids are versatile
and used in all phases of immunosuppression - induction therapy, maintenance therapy,
and treatment of acute rejection episodes. High doses are especially effective in reversing
acute rejection. Long-term use can result in hyperglycemia (high blood sugar),
osteoporosis (weakened bones), weight gain, cataracts, and increased susceptibility to
infections.
Examples: Prednisone, Methylprednisolone.
3 Antiproliferative Agents: These drugs inhibit the synthesis of nucleotides necessary for
lymphocyte (T and B cell) proliferation. Mycophenolate Mofetil (MMF) targets inosine
monophosphate dehydrogenase, a key enzyme in purine synthesis, selectively inhibiting
lymphocyte proliferation. Azathioprine acts as a purine analog, interfering with DNA
and RNA synthesis, broadly suppressing the proliferation of immune cells. Commonly
used as part of maintenance therapy alongside calcineurin inhibitors to provide additional
suppression of immune responses. Side effects include bone marrow suppression, leading
to reduced blood cell counts, gastrointestinal disturbances (nausea, diarrhea), and a higher
risk of infections.
Examples: Mycophenolate Mofetil (MMF), Azathioprine.
4 mTOR Inhibitors: mTOR inhibitors block the mammalian target of rapamycin (mTOR),
a protein kinase involved in cell growth, proliferation, and metabolism. By inhibiting
mTOR, these drugs suppress the activation and proliferation of T cells and other immune
cells. It is an alternative to calcineurin inhibitors, particularly useful for patients
experiencing nephrotoxicity from traditional regimens. They also promote anti-fibrotic
effects, reducing scarring in the transplanted organ. Delayed wound healing,
hyperlipidemia (high cholesterol levels), and increased susceptibility to infections are
common side effects. mTOR inhibitors require careful monitoring in post-operative
settings.
Examples: Sirolimus (Rapamycin), Everolimus.
5 Monoclonal Antibodies: Monoclonal antibodies are engineered to target specific
molecules on immune cells. Basiliximab and Daclizumab block the IL-2 receptor on T
cells, preventing their activation and proliferation. Rituximab targets CD20 on B cells,
depleting B cells and reducing antibody-mediated rejection. These antibodies are widely
used in induction therapy and the treatment of acute rejection cases, particularly in
antibody-mediated rejection (AMR). Side effects include infusion-related reactions,
increased infection risk, and potential allergic responses during administration.
Examples: Basiliximab, Daclizumab, Rituximab.
6 Polyclonal Antibodies: Polyclonal antibodies target multiple antigens on T cells and
other immune cells, leading to their depletion and suppression of immune responses.
ATG specifically depletes lymphocytes, while Alemtuzumab targets CD52, a surface
marker on lymphocytes. It is highly effective for induction therapy and treating severe
rejection cases. They are particularly useful in patients at high risk for rejection.
Associated with cytokine release syndrome, bone marrow suppression, and increased
infection risk.
Examples: Antithymocyte Globulin (ATG), Alemtuzumab.
7 Costimulation Blockers: Belatacept blocks the interaction between CD28 on T cells and
B7 on antigen-presenting cells (APCs), preventing the activation of T cells. This
innovative mechanism bypasses traditional pathways, reducing dependency on
calcineurin inhibitors. It is an alternative for maintenance therapy, particularly in patients
who cannot tolerate calcineurin inhibitors. Increased risk of infections and post-transplant
lymphoproliferative disorder (PTLD) are significant concerns.
Examples: Belatacept.
About the Author
Dr. Y. Shanti Prabha
Senior Lecturer, Department of Zoology
Dr. V.S. Krishna Government Degree and PG College (Autonomous), Visakhapatnam, India