0% found this document useful (0 votes)
6 views27 pages

Immunobiochemistry Assignment

The document provides a detailed explanation of complement fixation, a key mechanism in the immune system that involves the binding of complement proteins to antigen-antibody complexes, leading to pathogen elimination and inflammation. It outlines the structure and activation pathways of the complement system, including the classical, alternative, and lectin pathways, as well as the biological functions and diagnostic applications of complement fixation. The Complement Fixation Test (CFT) is described as a laboratory assay used to detect specific antibodies or antigens in patient serum by measuring complement activation.

Uploaded by

calebjprince017
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as DOCX, PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
6 views27 pages

Immunobiochemistry Assignment

The document provides a detailed explanation of complement fixation, a key mechanism in the immune system that involves the binding of complement proteins to antigen-antibody complexes, leading to pathogen elimination and inflammation. It outlines the structure and activation pathways of the complement system, including the classical, alternative, and lectin pathways, as well as the biological functions and diagnostic applications of complement fixation. The Complement Fixation Test (CFT) is described as a laboratory assay used to detect specific antibodies or antigens in patient serum by measuring complement activation.

Uploaded by

calebjprince017
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as DOCX, PDF, TXT or read online on Scribd
You are on page 1/ 27

BENUE STATE UNIVERSITY, MAKURDI

DEPARTMENT OF MEDICINE

COURSE:
BIOCHEMISTRY

MATRIC NUMBER:
BSU/MBBS/20/1977

COURSE TITLE:
IMMUNOLOGY

COURSE FACILITATOR:
DR. AMALI

QUESTIONS:
1. Explain in detail the complement fixation.
2. What is antigen and its role in disease condition?
QUESTION 1:
EXPLAIN IN DETAIL THE COMPLIMENT FIXATION
Introduction
The immune system is the body’s defense mechanism against pathogens, such as bacteria,
viruses, and parasites. Among its key players is the complement system, an essential component
of both innate and adaptive immunity. The complement system is a complex network of plasma
proteins and receptors that work together to eliminate pathogens, promote inflammation, and
clear cellular debris.
Definition of the Complement System
The complement system refers to a cascade of over 30 proteins, most of which are synthesized in
the liver and circulate in the blood in inactive forms. These proteins become activated in
response to infections or tissue damage, either through direct recognition of microbial surfaces or
via antibody interactions. Upon activation, they initiate a series of reactions that amplify the
immune response, resulting in pathogen destruction and enhanced immune cell activity.
Importance of Complement Fixation
Complement fixation is a central mechanism of the complement system, describing the binding
of complement proteins to antigen-antibody complexes. This process serves as a bridge between
innate and adaptive immunity by linking antibodies, which are specific to pathogens, with the
complement system, which amplifies and executes immune responses. Complement fixation
plays a crucial role in:
1. Elimination of Pathogens: By forming the membrane attack complex (MAC) that lyses
microbial cells.
2. Immune Clearance: Facilitating the removal of immune complexes from circulation,
preventing tissue damage.
3. Inflammation: Recruiting immune cells to the site of infection or injury.
The Complement System
The complement system is a cornerstone of immune defense, functioning as a cascade of
sequentially activated proteins that synergize with other immune components to protect the host.
Below is a detailed overview of its structure, activation pathways, and biological functions.
Key Components of the Complement System
1. Plasma Proteins:
• Complement proteins are designated C1 through C9, with additional regulatory and
functional molecules (e.g., Factor B, D, and H).
• These proteins are largely produced by hepatocytes, though macrophages and epithelial
cells can also contribute.
2. Inactive Precursors and Cascade Activation:
• In their inactive state, complement proteins exist as zymogens (enzymatic precursors).
• Activation involves enzymatic cleavage into fragments, such as C3 → C3a and C3b.
3. Regulatory Proteins:
• Complement activation is tightly regulated to prevent host tissue damage. Examples
include:
• Factor H: Inhibits the alternative pathway.
• Decay-Accelerating Factor (DAF): Disrupts C3 and C5 convertases.
• CD59 (Protectin): Blocks MAC formation on host cells.
Pathways of Activation

The complement system can be activated through three pathways, each with distinct triggers and
mechanisms:

1. Classical Pathway:

• Triggered by the binding of antibodies (IgG or IgM) to antigens on a pathogen's surface.

• C1q, a subunit of the C1 complex, recognizes and binds to the Fc region of antibodies,
initiating the cascade.

• This pathway underscores the link between adaptive immunity (antibodies) and innate
immunity (complement).
2. Alternative Pathway:

• Does not require antibodies for activation.

• Triggered directly by microbial surfaces, such as bacterial lipopolysaccharides or fungal


cell walls.

• C3 undergoes spontaneous hydrolysis to C3(H2O), leading to activation of downstream


components.

• This pathway provides a rapid response to pathogens, particularly during the early stages
of infection.

3. Lectin Pathway:

• Activated by mannose-binding lectin (MBL), a protein that binds to specific sugar


residues on the surfaces of pathogens.

• MBL-associated serine proteases (MASPs) activate complement components, mirroring the


classical pathway in downstream activation.
Biological Functions of the Complement System
1. Opsonization:

• Complement protein C3b binds to the surface of pathogens, acting as an "eat me" signal for
phagocytes, such as macrophages and neutrophils.

2. Lysis of Pathogens:

• The terminal complement components (C5b-C9) form the membrane attack complex (MAC).

• MAC creates pores in the microbial membrane, leading to cell lysis and death.
3. Chemotaxis and Inflammation:
• Complement fragments C3a, C4a, and C5a (known as anaphylatoxins) attract immune
cells to the site of infection.
• These fragments also promote vasodilation and increase vascular permeability, enhancing
immune cell access to infected tissues.
4. Immune Complex Clearance:
• Immune complexes are cleared by binding to C3b, which facilitates their removal by
phagocytes or transport to the spleen and liver.
Significance in Immunity
The complement system acts as a double-edged sword:
• Protective Roles: It provides robust protection against pathogens and facilitates immune
regulation.
• Pathological Roles: Dysregulation can lead to autoimmune diseases (e.g., systemic lupus
erythematosus) or chronic inflammation (e.g., age-related macular degeneration).
Complement Fixation
Complement fixation is a critical mechanism in the immune system that facilitates the activation
of the complement cascade through the interaction between antibodies and antigens. The process
involves several complex biochemical reactions and plays a pivotal role in the immune defense
against pathogens. Here’s a detailed breakdown of complement fixation:
Definition of Complement Fixation
Complement fixation refers to the process in which complement proteins are fixed or bound to
an antigen-antibody complex. This leads to the activation of the complement system, triggering a
cascade of immune responses that help eliminate pathogens, clear immune complexes, and
induce inflammation. Complement fixation can be divided into multiple steps:
1. Antigen-Antibody Complex Formation: Antibodies bind to antigens on pathogen
surfaces or infected cells.
2. Complement Activation: The binding of the antibody-antigen complex triggers the
complement proteins to bind and activate in a cascade-like manner.
3. Resulting Immune Functions: These include pathogen destruction (lysis), enhanced
phagocytosis (opsonization), and inflammation.
Mechanism of Action
The process of complement fixation can be divided into the following stages:
Step 1: Antigen-Antibody Complex Formation
o The first step in complement fixation is the binding of antibodies to specific antigens
present on the surface of pathogens (bacteria, viruses, fungi) or infected cells.
o IgM antibodies are the most efficient initiators of complement fixation due to their
pentameric structure, which can bind multiple C1 molecules simultaneously. This multi-
binding capability increases the efficiency of complement activation.
o IgG, though less effective than IgM, can also initiate the classical pathway of
complement activation by binding to its specific antigen.
Step 2: Activation of the Classical Pathway
o The classical pathway is the primary route for complement fixation and is activated when
the C1 complex binds to the Fc region of an antibody that is complexed with an antigen.
o The C1 complex consists of three subunits: C1q, C1r, and C1s. Upon binding to the
antibody, C1q initiates the activation of C1r and C1s.
o This cascade activation results in the cleavage of the C4 and C2 proteins, forming a C3
convertase complex (C4b2a) that is capable of cleaving the central complement protein
C3 into its active fragments C3a and C3b.
o C3a acts as an anaphylatoxin, promoting inflammation, while C3b binds to the pathogen
surface, marking it for destruction (opsonization).
Step 3: Formation of C3 Convertase and Amplification
o The C3 convertase (C4b2a) cleaves C3 into C3a and C3b.
o C3b binds to the pathogen or immune complex, enhancing opsonization and facilitating
the clearance of pathogens by phagocytes.
o C3a and C5a serve as anaphylatoxins that induce local inflammation, increasing vascular
permeability, and attracting immune cells (chemotaxis). These molecules recruit
phagocytes such as neutrophils and macrophages to the site of infection.
Step 4: Formation of the Membrane Attack Complex (MAC)
o The terminal complement components (C5b, C6, C7, C8, and multiple C9 molecules)
assemble on the surface of the pathogen, forming the Membrane Attack Complex
(MAC).
o The MAC creates pores in the membrane of the target cell, disrupting its integrity,
leading to water influx, lysis, and eventual death of the pathogen.
o This process is particularly effective against gram-negative bacteria, which have thin
cell walls that make them more vulnerable to the MAC.
Classical Pathway of Compliment Fixation
Step 1: Antigen-Antibody Complex Formation
The classical pathway is initiated when specific antibodies, predominantly IgM or IgG, bind to
antigens on the surface of a pathogen or other target. This interaction results in the formation of
antigen-antibody complexes, exposing the Fc region of the antibody. The exposed Fc region is
critical for complement activation, as it provides a binding site for the C1 complex.
Step 2: Activation of C1 Complex
The C1 complex is the first component of the classical pathway and consists of three subunits:
• C1q: The recognition subunit that binds to the Fc region of antibodies.
• C1r and C1s: Protease subunits that become sequentially activated upon C1q binding.

C1q requires simultaneous binding to at least two Fc regions of antibodies to undergo


conformational changes. This activates C1r, which subsequently cleaves and activates C1s,
setting the stage for the next steps in the cascade.

Step 3: Cleavage of C4 and C2

Activated C1s cleaves the complement protein C4 into two fragments:

• C4a: A small anaphylatoxin that contributes to inflammation.


• C4b: A larger fragment that binds covalently to the pathogen surface or immune
complex, creating a docking site for C2.

C2 is then cleaved by C1s into:

• C2a: A functional fragment that forms part of the C3 convertase.


• C2b: A smaller byproduct released into the fluid phase.

The binding of C4b and C2a forms the C3 convertase complex (C4b2a).

Step 4: Formation of C3 Convertase

C3 convertase (C4b2a) is a pivotal enzyme in the complement cascade. Its primary role is to
cleave the abundant complement protein C3 into:

• C3a: An anaphylatoxin that promotes inflammation by recruiting immune cells and


increasing vascular permeability.
• C3b: A key opsonin that tags pathogens for phagocytosis and also contributes to the
formation of C5 convertase.

The amplification potential of this step is significant, as one C3 convertase can cleave hundreds
of C3 molecules.

Step 5: Formation of C5 Convertase

When additional C3b molecules associate with the C3 convertase, the complex transitions into
C5 convertase (C4b2a3b). This enzyme cleaves C5 into:

• C5a: A potent anaphylatoxin and chemoattractant that activates immune cells and
promotes inflammation.
• C5b: The initiating fragment for the terminal complement pathway.

Step 6: Terminal Complement Pathway and Membrane Attack Complex (MAC)

C5b binds sequentially to C6, C7, C8, and multiple C9 molecules to form the MAC. This
complex inserts into the membrane of the target cell, creating pores that disrupt osmotic balance
and lead to cell lysis. The MAC is especially effective against Gram-negative bacteria,
enveloped viruses, and other susceptible targets.
Conditions for Complement Fixation

 Antigen-Antibody Binding: Complement fixation only occurs when an antibody


specifically binds to its cognate antigen. This is essential for the classical pathway
activation.
 Calcium and Magnesium Ions: Complement fixation requires the presence of calcium
(Ca²⁺) and magnesium (Mg²⁺) ions, which are crucial for the activation of the C1
complex and the stability of the complement cascade.
 Specificity of Antibody Classes: IgM and IgG are the main antibodies that mediate
complement fixation. IgM is the most potent in complement activation due to its
structure, but IgG also plays a significant role, especially in chronic infections.

Outcomes of Complement Fixation


1. Direct Lysis of Pathogens:
The ultimate result of complement fixation is the formation of the Membrane Attack
Complex (MAC), which disrupts the pathogen’s membrane, causing osmotic lysis and
cell death. This is especially important in the defense against bacteria and viruses.
2. Enhanced Phagocytosis (Opsonization):
The binding of C3b to the pathogen surface enhances its recognition and uptake by
phagocytes such as macrophages and neutrophils. This process, known as opsonization,
significantly improves the efficiency of pathogen clearance from the body.
3. Inflammation:
Complement fixation also leads to the release of anaphylatoxins (C3a, C5a), which
increase vascular permeability, recruit immune cells (e.g., neutrophils), and promote the
inflammatory response. This ensures that immune cells are rapidly mobilized to the site
of infection, promoting the resolution of the infection.
Diagnostic Applications of Complement Fixation
Complement fixation plays an essential role in diagnostic immunology. By leveraging the
principle of complement activation, researchers and clinicians can detect the presence of specific
antigens or antibodies in a patient's serum, aiding in the diagnosis of infections and autoimmune
diseases.
Complement Fixation Test (CFT)
The Complement Fixation Test (CFT) is a traditional laboratory assay that uses the
complement fixation process to detect specific antibodies or antigens in patient serum. It
measures the ability of a sample to fix complement in the presence of a specific antigen-antibody
complex.
Principle of the CFT
 Antigen-Antibody Reaction: The test begins by introducing a specific antigen into a
serum sample containing potential antibodies. If the antibody specific to the antigen is
present, an antigen-antibody complex forms, triggering complement activation
 Complement Consumption: Complement proteins are activated and fixed to the
immune complex. The test then determines whether complement was consumed during
the reaction.

 Hemolytic Indicator: After incubation, red blood cells coated with hemolysin (which
induces cell lysis in the presence of complement) are added.
o If complement was fixed by the antigen-antibody complex, no complement is
available to lyse the red blood cells.
o If no antigen-antibody complex formed (i.e., the antibody is absent), complement
remains unbound and lyses the red blood cells, producing a positive reaction.
Procedure of the CFT
1. Incubation of Antigen and Serum: A known antigen is mixed with the patient’s serum
to allow the formation of antigen-antibody complexes.
2. Complement Addition: Complement proteins (either from the patient's serum or a
standardized source) are added to the mixture.
3. Hemolysin and Red Blood Cells: Sheep red blood cells coated with hemolysin are
introduced.
4. Observation: If complement has been consumed, hemolysis is inhibited, and the result is
negative. If complement is unconsumed, hemolysis occurs, and the result is positive.

Complement Fixation Test


When antigen and antibody interact with each other, they form a complex called antigen-
antibody (Ag-Ab ) complex. The complex then interacts with complement protein and
gets fixed with it. After fixing, the complement degrades or gets cleaved into two
fragments i.e. smaller and larger fragments.
For eg. C2 gets fragmented into C2a and C2b. The larger fragments or active sites remain
attached to the Ag-Ab complex whereas smaller fragments separate and act as the
signaling molecule. The signaling molecule provides the signal to macrophages for
engulfment of complex and destruction of the antigen. It is a biological or in vivo
mechanism.
The complement fixation test Is based on the principle that the Ag-Ab complex can only
fix the complement and its effect on the hemolysis of RBC used in the indicator system.
If the sample contains desired antibodies or antigens, the Ag-Ab complex will be formed
in the sample after the addition of a complementary reactant(antigen or antibody, based
on the component being detected), and the indicator system will not be able to react to the
added complement(as it already gets fixed with Ag-Ab complex) which results in no
change in the indicator system.
No change in the indicator system refers to no lysis of RBC or no hemolysis.
Positive test
Antibody in sample + Antigen (added) + Complement → Ag-Ab Complex Fixed
with Complement
Complement fixed Ag-Ab + Indicator System → No change (No hemolysis)
Negative test
Sample with no antibody + Antigen (added) + Complement → Free Complement
Antigen (added) + Antibody in indicator system (On RBC) → Ag-Ab complex
Ag-Ab complex + Complement → Fixed Complement System → Hemolysis
Clinical Relevance and Applications
1. Infection Diagnosis:
CFT is used to detect antibodies against pathogens such as bacteria (e.g., Treponema
pallidum in syphilis), viruses (e.g., Influenza, Hepatitis), and parasites. This allows for
the identification of infections in early or latent stages, as antibodies specific to the
pathogen are often detectable before the appearance of clinical symptoms.
2. Autoimmune Disease Detection:
CFT is also useful in diagnosing autoimmune diseases where the immune system
mistakenly targets the body’s own cells. For instance, Systemic Lupus Erythematosus
(SLE), which involves immune complexes, can be identified by complement activation.
3. Blood Typing:
The CFT has been applied in blood typing to determine compatibility between donor and
recipient blood. This is particularly important in transfusions to prevent hemolytic
reactions.
4. Testing for Hypersensitivity:
CFT can be used to investigate hypersensitivity reactions such as delayed-type
hypersensitivity (DTH), where complement activation contributes to tissue damage.
Sensitivity and Specificity of the CFT
 Sensitivity: The CFT is highly sensitive because even low levels of specific antibodies
can trigger complement activation. It is capable of detecting infections or autoimmune
responses that are not immediately evident through clinical examination.
 Specificity: The specificity of the test relies on the proper selection of antigens, quality
control of reagents, and correct interpretation of hemolysis patterns. False positives or
negatives can occur due to sample handling errors, cross-reactivity, or the presence of
interfering substances in the patient’s serum.
Implications and Limitations of Complement Fixation
The complement fixation process is foundational to both immune defense mechanisms and
diagnostic techniques in immunology. While it has tremendous utility in these areas, there are
several implications and inherent limitations that must be addressed to fully understand its scope
and constraints.
Implications of Complement Fixation
1. Immune System Functionality
o Efficient Pathogen Clearance: Complement fixation enhances the immune
system's ability to target and eliminate pathogens through lysis, phagocytosis, and
inflammation.
o Defense Against a Broad Range of Microbes: It is effective against bacteria,
viruses, and some parasites, offering a versatile immune response.
o Immunological Homeostasis: Complement fixation helps remove apoptotic cells
and immune complexes, reducing the risk of chronic inflammation and tissue
damage.
2. Disease Pathogenesis
o Autoimmune Disorders: Overactivation of the complement system can lead to
tissue damage in autoimmune diseases such as systemic lupus erythematosus
(SLE) and rheumatoid arthritis (RA).
o Chronic Inflammation: Complement fixation may contribute to chronic
inflammatory diseases, including asthma and inflammatory bowel disease (IBD).
o Pathogen Evasion: Certain pathogens, such as Neisseria gonorrhoeae and
Staphylococcus aureus, have evolved mechanisms to evade complement-
mediated lysis, complicating immune clearance.
3. Therapeutic Potential
o Targeting Complement Proteins: Therapeutics that modulate complement
activity are emerging as treatments for autoimmune diseases and complement-
related disorders. For example, Eculizumab, a monoclonal antibody against C5,
is used to treat paroxysmal nocturnal hemoglobinuria (PNH) and atypical
hemolytic uremic syndrome (aHUS).
o Vaccine Design: Understanding complement fixation mechanisms aids in the
development of vaccines that enhance antibody-mediated complement activation,
boosting immune protection.
4. Diagnostic Innovations
o Complement fixation tests (CFT) have been historically significant in diagnosing
infectious diseases and autoimmune conditions. Though largely replaced by
modern techniques, their principles form the foundation of current immunological
assays.
o Advances in complement-based diagnostics, such as assays measuring
complement split products (e.g., C3a, C5a), have provided deeper insights into
disease states and immune response
Limitations of Complement Fixation in Immunity
1. Dependence on Specific Conditions
o Calcium and Magnesium Ions: The process requires these ions, making it
susceptible to inhibition in conditions where ion levels are altered.
o Optimal pH and Temperature: Complement proteins function efficiently only
under specific physiological conditions, limiting their activity in certain
microenvironments.
2. Pathogen Evasion Mechanisms
o Capsule Formation: Pathogens such as Streptococcus pneumoniae produce
capsules that inhibit complement deposition.
o Complement-Regulating Proteins: Viruses like HIV and bacteria like Borrelia
burgdorferi produce proteins that inactivate complement components, evading
immune detection.
o Biofilm Formation: Some bacteria form biofilms, creating a physical barrier that
reduces the efficiency of complement fixation.
3. Tissue Damage
o Excessive complement activation can result in bystander damage to host tissues.
This is evident in diseases like sepsis, where complement overactivation
exacerbates systemic inflammation.
4. Limited Role in Intracellular Pathogens
o Complement fixation primarily targets extracellular pathogens. Intracellular
pathogens such as Mycobacterium tuberculosis and certain viruses evade
complement-mediated destruction by residing within host cells.
Limitations of Complement Fixation Test (CFT)
1. Sensitivity and Specificity Issues
o The CFT relies heavily on high-quality reagents and proper sample handling.
Small errors can lead to false positives or negatives, reducing its reliability.
o Cross-reactivity with non-target antigens can lead to misinterpretation of results.
For instance, antibodies against unrelated pathogens may produce false-positive
results due to nonspecific binding.

2. Inability to Quantify Antibodies


o The CFT is qualitative rather than quantitative, providing a binary result
(positive/negative). Modern diagnostic tests such as ELISA or Western blot are
preferred for quantifying antibody levels.
3. Time-Consuming Procedure
o The CFT requires several steps, including incubation and observation, making it
slower compared to newer diagnostic techniques such as PCR or rapid antigen
tests.
4. Replacement by Modern Techniques
o While historically significant, the CFT has been largely replaced by more accurate
and efficient assays such as enzyme-linked immunosorbent assay (ELISA),
flow cytometry, and nucleic acid-based diagnostics.

QUESTION 2:
WHAT IS ANTIGEN AND IT’S ROLE IN DISEASE CONDITION
1. Introduction
An antigen is any substance that elicits an immune response when introduced into the body.
Derived from the term “antibody generator,” an antigen is recognized by the immune system as
either “self” (a normal component of the body) or “non-self” (foreign material). The primary role
of antigens is to enable the immune system to distinguish between these two categories, allowing
for the defense against harmful entities while preserving the body’s own tissues.
Antigens are crucial in both health and disease conditions. In the context of pathogens, antigens
are typically molecules found on the surface of bacteria, viruses, fungi, or other harmful
microorganisms. When these foreign substances invade the body, the immune system identifies
them as threats and mounts a response to neutralize or eliminate them. This process involves
both innate immunity (nonspecific defense mechanisms) and adaptive immunity (specific,
memory-based responses).
Apart from their role in infections, antigens also have critical implications in autoimmune
diseases, organ transplantation, allergies, and even cancer. In autoimmune conditions, for
example, the immune system fails to differentiate between self and non-self-antigens, leading to
attacks on the body’s own cells. Similarly, in organ transplantation, differences in antigens
between donor and recipient can trigger rejection. Understanding the molecular nature of
antigens is therefore vital for developing strategies to manage and treat various disease
conditions.
2. Structure of Antigens
Antigens exhibit a wide variety of chemical and structural characteristics, allowing them to
interact specifically with the immune system. These properties include their chemical
composition, structural complexity, and regions known as epitope s, which are recognized by immune

cells.

Chemical Composition
Antigens are typically macromolecules, with their composition determining their
immunogenicity (ability to provoke an immune response). The major types of antigens include:
1. Proteins: These are the most immunogenic antigens and are commonly found on the
surfaces of pathogens. Examples include viral envelope proteins and bacterial exotoxins.
o Example: Hemagglutinin on the influenza virus.
2. Polysaccharides: Found on bacterial cell walls, they often elicit strong immune
responses, particularly in conjunction with proteins.
o Example: Capsular polysaccharides in Streptococcus pneumoniae.
3. Lipids and Glycolipids: While not inherently immunogenic, they can become antigens
when associated with proteins or presented by specialized immune cells (e.g., via CD1
molecules).
o Example: Mycolic acids in Mycobacterium tuberculosis.
4. Nucleic Acids: These are weakly immunogenic on their own but can elicit responses in
specific contexts, such as during autoimmune conditions (e.g., anti-DNA antibodies in
lupus).
Epitopes
Epitopes are specific regions of an antigen that are recognized by immune cells, such as B-cell
receptors (BCRs) or T-cell receptors (TCRs). Each antigen may contain multiple epitopes,
enabling different immune cells to target the same antigen simultaneously. Epitopes can be
categorized as:
 Linear Epitopes: Formed by a continuous sequence of amino acids in a protein.
 Conformational Epitopes: Formed by the three-dimensional folding of a protein, where
amino acids from different parts of the sequence come together spatially.
The specificity of the immune response is determined by the interaction between epitopes and
immune receptors. Antigen-antibody binding, for instance, is highly specific and depends on the
structural compatibility between the epitope and the antibody.
Factors Influencing Antigenicity
Several factors determine how effectively an antigen can elicit an immune response:
1. Foreignness: The greater the difference between the antigen and the host’s own
molecules, the stronger the immune response.
2. Molecular Size: Larger molecules (greater than 10 kDa) are more likely to be
immunogenic.
3. Complexity: Antigens with diverse and complex structures, such as proteins with
multiple epitopes, are more immunogenic than simple molecules.
4. Accessibility: Epitopes must be accessible to immune cells or antibodies to induce a
response. Antigens buried within cell membranes or structures may evade detection.
Haptens
Haptens are small molecules that, on their own, cannot elicit an immune response. However,
when they bind to larger carrier proteins, they become immunogenic. This concept is important
in drug-induced allergies, where small drug molecules act as haptens and trigger immune
reactions upon binding to host proteins. For example, penicillin can act as a hapten, leading to
allergic responses in some individuals.
3. Types of Antigens
Antigens are molecules that can induce an immune response, and they vary greatly depending on
their origin, structure, and the way they interact with the immune system. There are three
primary categories of antigens: Exogenous Antigens, Endogenous Antigens, and Autoantigens.
Exogenous Antigens
Exogenous antigens are derived from external sources such as pathogens (bacteria, viruses,
fungi, and parasites) or toxins. These antigens are introduced into the body from the outside
environment and can be encountered through different entry points like the skin, respiratory
tract, gastrointestinal tract, or mucosal membranes.
Examples
Bacterial Antigens:
Bacteria often present a wide variety of antigens that trigger immune responses. For instance, the
lipopolysaccharides (LPS) found on the outer membrane of Gram-negative bacteria can act as
potent antigens, stimulating both innate and adaptive immune responses.
Viral Antigens:
Viruses produce proteins during their replication cycle that serve as antigens. For example, the
gp120 protein of the HIV virus or the hemagglutinin (HA) protein of the influenza virus are
recognized by the immune system as foreign antigens.
Fungal and Parasitic Antigens:
Antigens from fungi and parasites, such as the Candida albicans cell wall proteins or
Plasmodium falciparum antigens in malaria, also induce immune responses.
Toxins:
Many bacterial pathogens produce exotoxins (e.g., botulinum toxin or diphtheria toxin), which
act as antigens, causing severe immune reactions when introduced to the host.
Immune Response to Exogenous Antigens
These antigens are recognized by antigen-presenting cells (APCs) like dendritic cells,
macrophages, and B-cells. APCs engulf the pathogen or toxin through phagocytosis or
endocytosis and break it down into smaller peptides.
These peptides are then displayed on the surface of the APCs by Major Histocompatibility
Complex (MHC) Class II molecules.
-Helper T-cells (CD4+ T-cells) recognize these antigens and become activated, helping to
coordinate a broad immune response by stimulating B-cells to produce antibodies, activating
cytotoxic T-cells (CD8+ T-cells), and promoting inflammation.
Endogenous Antigens
Endogenous antigens are those that originate from within the host’s cells. These antigens can
result from intracellular pathogens like viruses or from cellular dysfunctions, such as
tumorigenesis. These antigens are often associated with abnormalities inside the body’s own
cells.
Examples
Viral Antigens:
When viruses infect host cells, they hijack the cell’s machinery to replicate and produce viral
proteins. These viral proteins, such as those found in influenza or hepatitis C infections, become
processed and presented as antigens by the host cell.
Tumor Antigens:
Cancerous cells can produce mutated or abnormal proteins (tumor-associated antigens) that the
immune system can recognize as foreign. For instance, the HER2 protein overexpressed in breast
cancer cells or p53 mutations in various cancers can act as endogenous antigens.
Cellular Malfunction:
Even normal cellular processes can generate antigens. For example, heat shock proteins or
altered peptides that emerge due to stress or malfunction can trigger immune recognition.
Immune Response to Endogenous Antigens
- MHC Class I molecules are responsible for presenting these endogenous antigens. These
molecules are present on all nucleated cells.
- Cytotoxic T-cells (CD8+ T-cells) are the primary effectors in responding to endogenous
antigens. They recognize antigenic peptides presented by MHC Class I molecules on infected or
cancerous cells and are activated to kill the target cell directly via the release of perforins and
granzymes.
Autoantigens
Autoantigens are components of the body’s own cells that are normally tolerated by the immune
system. However, in certain circumstances, the immune system mistakenly recognizes these self-
antigens as foreign, leading to autoimmune diseases. The failure of the immune system to
distinguish self from non-self is termed autoimmunity.
Examples
Type 1 Diabetes:
In this condition, the immune system attacks pancreatic β-cells that produce insulin. glutamic
acid decarboxylase (GAD) and insulin are some of the autoantigens targeted by the immune
system.
Rheumatoid Arthritis:
This disease is characterized by the immune system attacking the synovial joints. Autoantigens
such as citrullinated proteins (e.g., filaggrin) are recognized as foreign.
Multiple Sclerosis:
In this neurological disorder, myelin (the fatty sheath around nerves) becomes an autoantigen,
leading to demyelination.
Immune Response to Autoantigens
- The immune system typically avoids attacking autoantigens through central tolerance (during
immune cell development in the thymus) and peripheral tolerance (in the periphery). However, a
breakdown in these tolerance mechanisms can lead to the activation of autoreactive T-cells and
the production of autoantibodies by B-cells.
- Autoantigens are processed and presented to T-cells, causing the immune system to launch an
attack against the body’s own tissues, leading to inflammation, tissue damage, and chronic
disease.
4. Antigen Recognition and Immune Response
The immune system employs sophisticated mechanisms to detect and respond to antigens. This
involves antigen recognition, activation of the immune system, and memory formation. The
adaptive immune response, which is highly specific and long-lasting, plays a central role in
defending against pathogens, controlling cancer, and maintaining tolerance to self-antigens.
Detection of Antigens by Immune Cells
- Antigen-Presenting Cells (APCs):
APCs, such as dendritic cells, macrophages, and B-cells, are crucial for the initial detection and
processing of antigens. These cells capture antigens from pathogens, apoptotic cells, or toxins
through phagocytosis or receptor-mediated endocytosis.
Once internalized, antigens are broken down into smaller peptide fragments, which are then
loaded onto MHC molecules and displayed on the surface of the APC.
Dendritic cells are especially important in initiating the adaptive immune response as they
migrate from the site of infection to lymph nodes, where they activate naïve T-cells.
- MHC Molecules:
MHC Class I molecules present endogenous antigens (e.g., viral peptides) to cytotoxic T-cells
(CD8+ T-cells), leading to the destruction of infected cells.
MHC Class II molecules present exogenous antigens (e.g., bacterial peptides) to helper T-cells
(CD4+ T-cells), activating a broader immune response, including the stimulation of B-cells and
macrophages.
Activation of Adaptive Immunity
- Helper T-cells (CD4+):
Upon recognizing antigens presented by MHC Class II on APCs, helper T-cells become
activated and differentiate into various subsets (e.g., Th1, Th2, Th17) that release cytokines to
orchestrate the immune response.
Cytokines secreted by helper T-cells can stimulate B-cells to produce antibodies, macrophages
to increase phagocytosis, and cytotoxic T-cells to perform their effector functions.
-Cytotoxic T-cells (CD8+):
These T-cells recognize antigens presented by MHC Class I molecules on infected or abnormal
cells. Once activated, cytotoxic T-cells destroy the target cells directly through perforin-mediated
cell lysis and granzyme-induced apoptosis.
This is a critical defense mechanism against viruses and tumors, where infected or cancerous
cells need to be eliminate
- B-cells and Antibodies
B-cells recognize antigens through their B-cell receptors (BCRs), which are membrane-bound
antibodies. Upon encountering an antigen, B-cells can differentiate into plasma cells that secrete
large amounts of antibodies specific to that antigen.
Antibodies neutralize pathogens by binding to them and preventing their ability to infect host
cells. They also promote pathogen elimination through mechanisms like opsonization (coating
pathogens for phagocytosis) and complement activation (leading to pathogen lysis).
Immune Memory
After the immune response to an infection or vaccination, memory T-cells and memory B-cells
are formed. These cells “remember” the antigen encountered and remain in circulation.
Upon subsequent exposure to the same antigen, the immune system can recognize the infection
and fight efficiently. Immunological memory is the ability of the immune system to respond
more rapidly and effectively to pathogens that have been encountered previously, and reflects the
preexistence of a clonally expanded population of antigen-specific lymphocytes.
Memory responses, which are called secondary, tertiary, and so on, depending on the number of
exposures to antigen, also differ qualitatively from primary responses. This is particularly clear
in the case of the antibody response, where the characteristics of antibodies produced in
secondary and subsequent responses are distinct from those produced in the primary response to
the same antigen. Memory T-cell responses have been harder to study, but can also be
distinguished from the responses of naïve or effector T cells.
5. Role of Antigens in Disease Conditions
Antigens play a crucial role in both the defense mechanisms of the immune system and the
pathogenesis of various diseases. The body’s ability to recognize antigens is integral to the
immune response, but when this recognition goes awry or is manipulated by pathogens or other
factors, it can result in disease. Below is an in-depth exploration of how antigens are involved in
infectious diseases, autoimmune diseases, cancer, and vaccination/immunotherapy.
Infectious Diseases
Infectious diseases are caused by pathogens such as bacteria, viruses, fungi, and parasites. These
pathogens often introduce foreign antigens into the body, which trigger the immune system to
respond and combat the infection. The antigens derived from these pathogens are recognized by
immune cells, and their presence is pivotal in both initiating immune responses and, in some
cases, causing disease symptoms.
Pathogen-Associated Antigens:
- Bacterial Antigens:
Bacteria have unique cell surface markers such as lipopolysaccharides (LPS) and peptidoglycan
that act as antigens. When these antigens are recognized by immune cells, they activate the
innate immune system (e.g., through toll-like receptors or TLRs) and then stimulate adaptive
immunity. Exotoxins produced by bacteria (e.g., botulinum toxin, diphtheria toxin) are also
potent antigens that can cause severe tissue damage, triggering inflammation and immune
responses.
- Viral Antigens:
Viruses express various proteins that are recognized by the immune system, such as capsid
proteins, envelope proteins, or viral enzymes. For instance, the gp120 protein of the HIV virus
binds to the immune cells, leading to their destruction and immune evasion. Similarly, viruses
like influenza and hepatitis C display viral proteins that are recognized by cytotoxic T-cells
(CD8+ T-cells). These viral antigens stimulate a strong immune response, but viruses can also
evolve mechanisms to evade immune detection, complicating effective immune clearance.
Immune Response to Infectious Antigens:
- Activation of T-cells and B-cells:
Upon encountering pathogen-derived antigens, T-helper cells (CD4+) are activated and stimulate
cytotoxic T-cells (CD8+) to destroy infected cells. Additionally, B-cells are activated to produce
specific antibodies targeting the pathogen. The production of antibodies neutralizes pathogens by
preventing their entry into host cells, and facilitates their clearance through processes such as
opsonization and complement activation.
- Pathogen Evasion:
Some pathogens can evade immune detection by antigenic variation (e.g., the influenza virus
changes its surface proteins frequently, making it hard for the immune system to recognize and
neutralize it). Additionally, some pathogens, like HIV, can downregulate MHC Class I
expression on infected cells, preventing antigen presentation and evading cytotoxic T-cell
recognition.

Autoimmune Diseases
In autoimmune diseases, the immune system mistakenly identifies self-antigens as foreign and
mounts an immune attack against the body’s own tissues. This occurs due to failures in immune
tolerance mechanisms, where autoreactive immune cells escape normal regulation and are able to
target normal, healthy cells as if they were pathogens. This misrecognition of self-antigens leads
to chronic inflammation, tissue damage, and the development of disease.
Mechanisms of Autoimmune Diseases:
- Loss of Tolerance:
Normally, the immune system is trained during development to tolerate the body’s own cells.
However, in autoimmune diseases, autoreactive T-cells and B-cells are activated and begin to
attack the body’s own tissues. This failure of central tolerance (in the thymus and bone marrow)
or peripheral tolerance (in the lymph nodes and other tissues) is often a key feature of
autoimmune diseases.
- Molecular Mimicry:
Sometimes, infections by certain pathogens can trigger autoimmune diseases due to molecular
mimicry, where the pathogen’s antigens resemble self-antigens. For example, a streptococcal
infection can trigger rheumatic fever, in which antibodies against the bacterial antigens cross-
react with heart tissue, leading to carditis.
Examples of Autoimmune Diseases:
- Type 1 Diabetes:
This disease occurs when the immune system attacks the pancreatic β-cells, which produce
insulin. The body’s own proteins, such as glutamic acid decarboxylase (GAD) and insulin itself,
are recognized as foreign antigens. This leads to the destruction of insulin-producing cells,
resulting in insulin deficiency and diabetes.
- Systemic Lupus Erythematosus (SLE):
In SLE, autoantibodies are produced against nuclear components, such as DNA, histones, and
ribonucleoproteins. These antibodies form immune complexes that can deposit in various tissues,
leading to inflammation and damage, particularly in the skin, kidneys, and joints.
- Rheumatoid Arthritis (RA):
In RA, the immune system targets joint tissues, and specific citrullinated peptides are recognized
as autoantigens. Rheumatoid factor (an antibody against IgG) and anti-citrullinated protein
antibodies (ACPA) are common biomarkers in RA.
Cancer
Cancer arises from the uncontrolled growth of abnormal cells. Tumor cells often present unique
or altered antigens, known as tumor-associated antigens (TAAs), that can be recognized by the
immune system. These antigens play a significant role in tumor detection and immune responses
to cancer.

Tumor-Associated Antigens (TAAs):


- Tumor-Specific Antigens (TSAs):
These are antigens that are exclusively expressed by tumor cells, often due to mutations in genes
encoding proteins that are not found on normal cells. For instance, mutations in the p53 gene, a
tumor suppressor gene, can lead to the production of mutated p53 proteins that act as TSAs.
- Oncofetal Antigens:
Some antigens, such as carcinoembryonic antigen (CEA) and alpha-fetoprotein (AFP), are
typically expressed during fetal development but can reappear in certain cancers, including
colorectal cancer (CEA) and liver cancer (AFP).
- HER2/neu:
Overexpression of the HER2 protein is common in breast cancer and is used as a diagnostic
marker and a target for therapy (e.g., trastuzumab, a monoclonal antibody, targets HER2 to
inhibit tumor growth).
Immune Response to Tumor Antigens:
- Immune Surveillance:
The immune system continuously monitors the body for abnormal cells, including cancer cells.
Cytotoxic T-cells (CD8+ T-cells) can recognize and destroy cancer cells presenting tumor
antigens via MHC Class I molecules. Natural Killer (NK) cells also contribute to the immune
response by recognizing and killing tumor cells in the absence of normal MHC Class I
expression.
- Immune Evasion by Tumors:
Tumor cells often develop strategies to evade immune recognition. For example, they may
downregulate MHC Class I expression, or they may produce immunosuppressive molecules like
PD-L1, which binds to PD-1 receptors on T-cells, effectively turning off the immune response.
-Cancer Immunotherapy:
Immunotherapies such as checkpoint inhibitors (e.g., anti-PD-1/PD-L1 and anti-CTLA-4
therapies) are designed to enhance the immune response against tumor antigens. Other therapies,
such as CAR-T cell therapy, involve engineering T-cells to recognize specific tumor antigens
and improve their ability to target and kill cancer cells.
Vaccination and Immunotherapy
Vaccination and immunotherapy are two critical medical interventions that exploit the immune
system’s ability to recognize antigens and mount a protective immune response.
- Vaccination:
Vaccines contain antigens derived from pathogens (such as inactivated viruses, bacterial
proteins, or toxoids) or synthetic versions of these antigens. These antigens stimulate the immune
system to generate memory T-cells and memory B-cells without causing disease. Upon
subsequent exposure to the pathogen, the immune system responds quickly and effectively.
Examples of vaccines include those for measles, polio, influenza, and HPV. mRNA vaccines
(e.g., for COVID-19) use messenger RNA to instruct cells to produce a harmless portion of the
pathogen (such as the spike protein of SARS-CoV-2), which then triggers an immune response.
- Immunotherapy:
Immunotherapy aims to boost or enhance the body’s immune system to fight diseases like
cancer. Tumor antigens are targeted using monoclonal antibodies or vaccines, allowing the
immune system to recognize and destroy tumor cells more effectively.
Monoclonal antibodies (e.g., rituximab for lymphoma) are laboratory-made antibodies that bind
to tumor antigens, facilitating their destruction through immune mechanisms such as antibody-
dependent cellular cytotoxicity (ADCC).
Cancer vaccines (e.g., Bacillus Calmette-Guérin (BCG) for bladder cancer) stimulate the
immune system to recognize and target specific tumor-associated antigens.
REFERENCES

Abbas, A. K., Lichtman, A. H., & Pillai, S. (2018). Basic Immunology: Functions and Disorders
of the Immune System. Elsevier.

Abbas, A. K., Lichtman, A. H., & Pillai, S. (2018). Basic Immunology: Functions and Disorders
of the Immune System (6th ed.). Elsevier.

Atkinson, M. A., Eisenbarth, G. S., & Michels, A. W. (2014). Type 1 diabetes. *The Lancet*,
383(9911), 69-82. https://doi.org/10.1016/S0140-6736(13)60591-7

Cohen, P. L., & Eisenberg, R. A. (2000). Autoimmunity to self: The genesis of systemic lupus
erythematosus. The New England Journal of Medicine, 343(16), 1157-1166.
https://doi.org/10.1056/NEJM200010193431607

Dawson, R., & Sakamoto, S. (2004). Antigens and the immune response. In Immunology: A
short course (3rd ed., pp. 22-40). Wiley-Liss.

Firestein, G. S. (2009). Evolving concepts of rheumatoid arthritis. *Nature*, 423(6937), 356-361

Janeway, C. A., Travers, P., Walport, M., & Shlomchik, M. J. (2001). Immunobiology: The
immune system in health and disease (5th ed.). Garland Publishing

Janeway, C. A., Travers, P., Walport, M., & Shlomchik, M. J. (2001). Immunobiology: The
Immune System in Health and Disease. Garland Science.

Lane, D. P. (1992). Cancer. P53, guardian of the genome. Nature, 358(6381), 15-16.
https://doi.org/10.1038/358015a0

Müller, M., Jassoy, C., & Kirchner, H. (2013). HIV immune evasion and immune activation.
*Annual Review of Immunology*, 11, 311-341.
https://doi.org/10.1146/annurev.immunol.11.1.311

Murphy, K., Weaver, C., & Berg, L. (2017). Janeway’s Immunobiology. Garland Science.

O’Connell, J. T., et al. (2003). Characterization of a new oncofetal antigen: Carcinoembryonic


antigen (CEA). Cancer Research, 63(13), 4879-4886.
Slamon, D. J., et al. (1987). Human breast cancer: Correlation of relapse and survival with
amplification of the HER-2/neu oncogene. Science, 235(4785), 177-182.
https://doi.org/10.1126/science.3798106

Swan, G. E. (1997). Molecular mimicry in autoimmune diseases. The Lancet, 349(9052), 1227-
1229. https://doi.org/10.1016/S0140-6736(96)11017-9

Walport, M. J. (2001). Complement. First of two parts. New England Journal of Medicine,
344(14), 1058–1066.

Walport, M. J. (2001). Complement. First of two parts. New England Journal of Medicine,
344(14), 1058–1066. https://doi.org/10.1056/NEJM200104053441406

Weaver, C. T., et al. (2018). Immunology: The immune system in health and disease (8 th ed.).
Elsevier.

Zinkernagel, R. M., & Doherty, P. C. (1974). Restriction of in vitro T cell-mediated cytotoxicity


in lymphocytic choriomeningitis involving the recognition of virus-infected cells by T
cells. Nature, 251(5475), 547-549. https://doi.org/10.1038/251547a0

You might also like