Immunology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 46

INTRODUCTION TO IMMUNOLOGY

List the main historical stages in the development of immunology.


The main historical stages in the development of immunology can be summarized as
follows:
​ Girolamo Fracastro (16th Century): He wrote about contagion as a potential cause
of diseases, laying the groundwork for the concept of infectious agents.
​ Edward Jenner (1798): Jenner introduced the concept of immunity in response to
contagion. He observed that individuals who had cowpox infections were resistant to
smallpox, which led to the development of vaccination.
​ Joseph Lister (19th Century): Lister introduced the concept of aseptic surgery,
emphasizing the importance of maintaining a sterile environment during medical
procedures.
​ Julius Bordet (late 19th Century): Bordet made significant contributions to
immunology by demonstrating the need for two factors, a specific sensitizer, and a
non-specific factor (Alexine), for processes like bacteriolysis and RBC lysis.
​ Louis Pasteur (19th Century): Pasteur contributed to immunology by introducing
the concept of attenuation, explaining how exposure to adverse conditions could alter
the virulence of pathogens. He also developed the first vaccines against cholera,
anthrax, and rabies.
​ Klebs and Loeffler (late 19th Century): They isolated the diphtheria bacillus, a
crucial step in understanding the causative agent of diphtheria.
​ Metchnikoff (late 19th Century): Metchnikoff, a Russian zoologist, introduced the
concept of cellular immunity by demonstrating phagocytosis of fungal spores by white
blood cells, highlighting the role of immune cells in defense.
​ Robert Koch (late 19th Century): Koch's research revealed that animals could be
immunized by exposure to Tubercle bacilli, which led to the understanding of immune
responses. He also described phenomena related to the response of the skin to
Tuberculous bacilli.
​ Julius Bordet (20th Century): Bordet ultimately discovered the complement system,
a fundamental part of the immune response, and was awarded the Nobel Prize.
​ Kraus (20th Century): Kraus discovered precipitation by mixing a patient's serum
and filtrate of typhoid bacilli, contributing to diagnostic immunology.
​ Landsteiner (20th Century): Landsteiner made significant contributions by
discovering blood group antigens and antibodies. He introduced the concept of
incomplete antigens (Haptens) and dominated the field of immunology for several
decades. He was also awarded the Nobel Prize.

Define the science of immunology.


Immunology is a relatively new branch of the medical sciences. It started as a branch of
microbiology, which led to the study of infectious diseases and then the body’s response to
them.
Define the subject of study of immunology.
The subject of study in immunology is the immune system and its various components,
functions, and responses. Immunology is the scientific discipline that investigates the
immune system's roles in defending the body against pathogens (such as bacteria, viruses,
and fungi), recognizing and eliminating abnormal or harmful cells (including cancer cells),
and maintaining overall health.
List the main areas of immunology: general immunology and private immunology.
​ General Immunology:
● General immunology refers to the foundational and comprehensive study of
the immune system. It encompasses the fundamental principles,
mechanisms, and components of immunity, as I explained in the previous
response. This includes innate and adaptive immunity, immune responses,
immune cells, antibodies, antigen recognition, and various aspects of
immunological science.
​ Specialized or Private Immunology:
● Private immunology, in this context, might refer to more specific or specialized
branches or subfields within immunology. These areas of study focus on
particular aspects or applications of immunology, and they may include:
● Clinical Immunology: Focused on the clinical aspects of
immune-related diseases and patient care, including autoimmune
disorders, immunodeficiencies, and allergic conditions.
● Infectious Disease Immunology: Concentrated on the interactions
between the immune system and infectious agents, such as bacteria,
viruses, fungi, and parasites.
● Cancer Immunology: Investigating the role of the immune system in
recognizing and targeting cancer cells, and the development of
immunotherapies for cancer treatment.
● Transplantation Immunology: Studying the immune responses
involved in organ transplantation and the prevention of graft rejection.
● Allergy and Immunology: Concerned with allergic reactions,
hypersensitivities, and immune responses to allergens.
● Vaccine Development: Focusing on the design and creation of
vaccines to induce protective immunity against specific diseases.
● Reproductive Immunology: Exploring immune-related aspects of
pregnancy, fetal development, and fertility.

Discuss the place of the discipline of immunology in individual specialties.


​ Allergy and Immunology: This specialty focuses on allergic diseases,
immunodeficiencies, and immune system disorders. Allergists and immunologists
diagnose and manage conditions like asthma, food allergies, and primary
immunodeficiencies.
​ Transplant Surgery: Transplant immunology is crucial for organ and tissue
transplantation. Transplant surgeons work closely with immunologists to assess
compatibility, manage graft rejection, and develop strategies for immune tolerance in
transplant recipients.
​ Infectious Disease Medicine: Immunology is central to infectious disease medicine,
as it helps in understanding how the immune system responds to various pathogens.
Immunologists and infectious disease specialists collaborate to diagnose and
manage infections, including HIV, tuberculosis, and emerging infectious diseases.
​ Oncology (Cancer Treatment): Cancer immunology is a rapidly growing field within
oncology. Immunotherapies, such as immune checkpoint inhibitors and CAR-T cell
therapy, harness the immune system to target and destroy cancer cells.
Immunologists and oncologists work together to develop and administer these
treatments.

Describe the task of general immunology.

The task of general immunology is to understand and explore the fundamental principles,
mechanisms, and components of the immune system. It encompasses a wide range of
topics related to how the immune system functions, including the following:
​ Immune System Components: General immunology examines the various cells,
proteins, and molecules that make up the immune system. This includes white blood
cells (leukocytes), such as T cells, B cells, macrophages, and neutrophils, as well as
antibodies and cytokines.
​ Innate and Adaptive Immunity: It delves into the concepts of innate immunity (the
body's immediate, non-specific defense mechanisms) and adaptive immunity (the
ability to develop specific responses to pathogens over time).
​ Immune Responses: The study of how the immune system recognizes and
responds to pathogens, such as bacteria, viruses, fungi, and parasites. This includes
mechanisms like phagocytosis, cytokine signaling, and the activation of immune
cells.
​ Antigen Recognition: Investigating how the immune system recognizes and
distinguishes between "self" and "non-self" substances through antigen recognition.
This includes the role of major histocompatibility complexes (MHC), T cell receptors,
and B cell receptors.
​ Immunological Memory: Understanding how the immune system forms memory
responses, allowing it to respond more effectively upon re-exposure to the same
pathogen. This is the basis of vaccination.
And more…

Innate immunity
1. List and discuss components of innate immunity.
Anatomical Defenses:
​ Skin: The body's largest organ serves as a physical barrier that prevents pathogens
from entering. The outermost layer of the skin is composed of dead cells and keratin,
which are inhospitable to most pathogens.
​ Mucous Membranes: These line the respiratory, gastrointestinal, and urogenital
tracts and secrete mucus, which traps pathogens and other foreign particles. Mucous
membranes are equipped with hair-like structures called cilia that help move trapped
particles out of the body.
Physiological Defenses:
​ Acidic Environments: Various bodily fluids, such as gastric acid in the stomach and
vaginal secretions, are acidic. The low pH in the stomach, for instance, helps destroy
ingested pathogens.
​ Enzymes: Enzymes like lysozyme, found in tears, saliva, and other secretions, can
break down the cell walls of certain bacteria.
​ Antimicrobial Peptides: These are small proteins with antimicrobial properties that
can directly kill or inhibit the growth of pathogens. For example, defensins are
antimicrobial peptides found in various bodily fluids.
Phagocytic Defenses:
​ Phagocytes: White blood cells known as phagocytes are key players in innate
immunity. Neutrophils and macrophages are examples of phagocytes. They engulf
and digest pathogens by a process called phagocytosis.
Inflammatory Defenses:
​ Inflammation: Innate immunity triggers an inflammatory response when it detects
infection or injury. The signs of inflammation include redness, heat, swelling, and pain
at the site of infection or injury.
● Inflammatory Mediators: Release of substances like histamines and
prostaglandins that promote inflammation.
● Increased Blood Flow: Dilation of blood vessels to enhance blood flow to
the affected area.
● Recruitment of Immune Cells: Chemotactic factors attract immune cells,
like neutrophils and macrophages, to the site of infection.
● Tissue Repair: Inflammation also initiates the repair of damaged tissues.

2. Justify external barriers against infection.


External barriers are crucial components of the body's innate immune system and play a
significant role in defending against infections. These barriers serve as the first line of
defense, providing immediate and non-specific protection. Here are some justifications for
the importance of external barriers against infection:
​ Preventing Pathogen Entry: The primary function of external barriers like the skin
and mucous membranes is to physically block pathogens from entering the body.
The intact skin and the epithelial cells lining the mucous membranes create a
formidable physical barrier that many microorganisms cannot breach.
​ Barrier Reinforcement: Skin is the body's largest organ and represents a robust
physical barrier. Its outermost layer consists of dead, keratinized cells that are highly
impermeable to pathogens. This makes it challenging for most microbes to penetrate.
​ Mucous Trapping: Mucous membranes in the respiratory, gastrointestinal, and
urogenital tracts produce mucus, a sticky substance that traps pathogens and other
foreign particles. The action of cilia helps move this trapped material away from
vulnerable tissues.
​ Acidic Environment: The stomach's acidic environment, with a low pH, is a
physiological barrier that can kill many ingested pathogens. This acidic environment
is hostile to most bacteria and microorganisms.
​ Infection Prevention: By preventing pathogen entry and colonization, external
barriers help avoid infections in the first place. This is a proactive and efficient way to
safeguard the body against various diseases.

3. Identify and classify cells of the immune system.


Immunity = Innate + adaptive immunity

Innate immunity cells: Phagocytes(dendritic cells, Mast cells, Macrophages), granulocytes


(neutrophils, eosinophils, basophils)
and natural killer (NK) cells
Adaptive immunity cells: B lymphocytes and T lymphocytes
4. Classify the types of pattern recognition receptors.
Pattern recognition receptors (PRRs) are a crucial part of the innate immune system, helping
to identify and respond to various pathogen-associated molecular patterns (PAMPs) present
on microorganisms. PRRs are categorized into several groups based on their structural and
functional characteristics:
Toll-Like Receptors (TLRs): TLRs are transmembrane proteins located on the cell surface
or within endosomes. They recognize a wide range of microbial components, including
lipopolysaccharides (LPS), lipoproteins, and nucleic acids such as double-stranded RNA.
TLR activation triggers an immune response.
Nucleotide-Binding Oligomerization Domain (NOD)-Like Receptors (NLRs): NLRs are
cytoplasmic receptors that recognize intracellular PAMPs or danger-associated molecular
patterns (DAMPs). They play a role in detecting bacterial components like peptidoglycan.
Activation of NLRs can lead to the formation of inflammasomes, initiating the release of
proinflammatory cytokines.
RIG-I-Like Receptors (RLRs): RLRs are another group of cytoplasmic receptors. They
recognize viral RNA, specifically single-stranded RNA molecules with 5'-triphosphate ends.
Activation of RLRs induces the production of type I interferons (IFNs).
C-type Lectin Receptors (CLRs): CLRs are membrane-bound or soluble receptors that
recognize carbohydrates on the surface of pathogens. They play a role in recognizing fungal
components like beta-glucans and mannans. CLRs can activate immune responses and
influence the adaptive immune system.
Scavenger Receptors: Scavenger receptors are typically expressed on macrophages and
are involved in the recognition and uptake of a broad range of microbial ligands, including
modified low-density lipoproteins.
5. Explain initiation of an immune response.
​ Recognition of Pathogens:
● Innate Immune System: The process usually starts with the innate immune
system. Innate immune cells, such as macrophages and dendritic cells, use
pattern recognition receptors (PRRs) to detect molecular patterns associated
with pathogens (PAMPs - Pathogen-Associated Molecular Patterns). PRRs
include Toll-like receptors (TLRs) and NOD-like receptors (NLRs).
● Adaptive Immune System: If the innate immune system cannot completely
eliminate the threat, it signals the adaptive immune system.
Antigen-presenting cells (APCs), mainly dendritic cells, capture and process
antigens (parts of the pathogen) to present them to T cells, which are key
players in the adaptive immune response.
​ Activation of Immune Cells:
● Innate Immune System: Upon detecting pathogens, innate immune cells
become activated. For example, macrophages and neutrophils are recruited
to the site of infection to engulf and destroy the invaders. Complement
proteins, which are part of the innate immune system, can also help
neutralize pathogens.
● Adaptive Immune System: In the adaptive immune system, T cells and B cells
become activated. T cells are activated by recognizing antigens presented by
APCs. Helper T cells assist in activating other immune cells, while cytotoxic T
cells directly target infected cells. B cells, on the other hand, produce
antibodies (immunoglobulins) that can neutralize pathogens.
​ Clonal Expansion:
● In both the adaptive and innate immune systems, activated immune cells
undergo clonal expansion. This means that once an immune cell is activated,
it multiplies rapidly to create a larger army of cells specific to the detected
pathogen.
​ Effector Phase:
● The expanded immune cells carry out effector functions. For example,
cytotoxic T cells target and destroy infected host cells, while B cells produce
antibodies that can bind to and neutralize pathogens. The innate immune
cells continue their attack on the invading pathogens.
​ Resolution:
● After the pathogen is eliminated or controlled, the immune response begins to
resolve. Some activated immune cells die off, while others become memory
cells.
​ Memory Cells:
● Both the adaptive and innate immune systems have memory components.
Memory cells "remember" the specific pathogen they encountered, allowing
for a faster and more effective response if the same pathogen is encountered
again in the future.

6. Know how do the innate immune system instigates adaptive immunity .


​ Recognition of Pathogens: The innate immune system is the first line of defense
and quickly recognizes the presence of pathogens through pattern recognition
receptors (PRRs). These receptors, such as Toll-like receptors (TLRs) and NOD-like
receptors (NLRs), detect molecular patterns associated with pathogens (PAMPs -
Pathogen-Associated Molecular Patterns).
​ Antigen Presentation: When innate immune cells, such as dendritic cells or
macrophages, detect a pathogen through PRRs, they phagocytose (engulf) the
pathogen. Within the phagocyte, the pathogen is broken down into smaller pieces,
and specific fragments (antigens) are processed and displayed on the cell's surface.
​ Activation of Adaptive Immune Cells: Antigen-presenting cells (APCs), particularly
dendritic cells, migrate to the lymph nodes, which are central hubs for immune
responses. There, they present the processed antigens to T cells and B cells.
​ T Cell Activation: Dendritic cells present antigens to helper T cells (CD4+ T cells). If
the presented antigen matches the T cell's specific receptor (T cell receptor or TCR)
and is accompanied by co-stimulatory signals, the T cell becomes activated.
​ B Cell Activation: In parallel, B cells recognize antigens directly, thanks to their B
cell receptors. Activation of B cells often requires help from helper T cells. B cells
then become activated and can differentiate into plasma cells, which produce
antibodies specific to the antigen.
​ Clonal Expansion: Activated T cells and B cells undergo clonal expansion, meaning
they multiply to form larger populations of immune cells specific to the pathogen.
​ Immune Response: The activated T cells can help regulate and coordinate the
immune response. Helper T cells stimulate cytotoxic T cells, which directly target and
destroy infected host cells, as well as B cells, which produce antibodies. Antibodies
produced by B cells neutralize the pathogen and assist in its removal.
​ Memory Formation: Once the immune response is successful, some T and B cells
become memory cells. These memory cells "remember" the specific pathogen and
provide immunological memory. If the same pathogen is encountered in the future,
the immune system can mount a faster and more effective response.

7. Explain collaboration between innate and adaptive immune responses.


The collaboration between innate and adaptive immune responses is essential for effective
protection against infections. These two branches of the immune system work together in a
coordinated manner to recognize, target, and eliminate pathogens. Here's how their
collaboration occurs:
​ Recognition of Pathogens: The innate immune system serves as the first line of
defense and rapidly detects the presence of pathogens through pattern recognition
receptors (PRRs), such as Toll-like receptors (TLRs). These receptors recognize
pathogen-associated molecular patterns (PAMPs) on the surface of microorganisms.
​ Inflammatory Response: Upon detecting a pathogen, innate immune cells, such as
macrophages and neutrophils, are recruited to the site of infection. They release
pro-inflammatory cytokines and chemokines, which help amplify the immune
response and attract more immune cells to the infection site.
​ Antigen Presentation: Innate immune cells, particularly dendritic cells, phagocytose
pathogens and process them. During this process, they present antigens derived
from the pathogen on their cell surface. This presentation is crucial for the initiation of
the adaptive immune response.
​ Activation of Adaptive Immunity: Antigen-presenting cells (APCs), especially
dendritic cells, migrate to secondary lymphoid organs, such as lymph nodes. There,
they present the processed antigens to helper T cells (CD4+ T cells) and interact with
other immune cells.
​ Helper T Cell Activation: Dendritic cells activate helper T cells by presenting
antigens along with co-stimulatory signals. Helper T cells, in turn, play a central role
in activating both cellular and humoral immune responses.
​ Cytokine Signaling: Helper T cells release cytokines that further activate and guide
various immune cells. For example, they can stimulate cytotoxic T cells, which
directly target and destroy infected cells, or they can help B cells produce antibodies.
​ B Cell Activation: B cells directly recognize antigens and, with the assistance of
helper T cells, become activated. They differentiate into plasma cells that secrete
antibodies specific to the pathogen.
​ Enhanced Innate Immunity: The adaptive immune response can also enhance the
effectiveness of the innate immune response. For example, antibodies produced by
B cells can neutralize pathogens and make them more susceptible to phagocytosis
by innate immune cells.
​ Immunological Memory: Both adaptive and innate immune responses contribute to
immunological memory. Memory T and B cells, as well as some innate immune cells,
"remember" the specific pathogen. If the same pathogen is encountered again, the
immune system can respond more rapidly and effectively.

8. Draw and describe interaction between innate and adaptive immune responses.

Adaptive immunity
1. List and describe the central and peripheral organs of the immune system.
Central Organs:
​ Bone Marrow: The bone marrow is a central organ where various immune cells are
produced. It is responsible for the generation of all blood cells, including white blood
cells (leukocytes), which are critical components of the immune system.
​ Thymus: The thymus is a gland located in the upper chest. It is a primary central
organ of the adaptive immune system, primarily involved in the development and
maturation of T cells (T lymphocytes). T cells are essential for cell-mediated immunity
and help orchestrate immune responses.
Peripheral Organs and Tissues:
​ Spleen: The spleen is a peripheral organ located under the ribcage. It acts as a
blood filter and immune organ. It contains white pulp, where immune responses
against blood-borne pathogens take place. The spleen filters blood and removes
damaged red blood cells and foreign materials.
​ Lymph Nodes: Lymph nodes are small, bean-shaped structures found throughout
the body, connected by the lymphatic system. They filter lymph, a clear fluid
containing immune cells and cellular debris. Lymph nodes are essential for detecting
and responding to infections. They contain B cells, T cells, and other immune cells.
​ Lymphatic System: The lymphatic system comprises a network of vessels, nodes,
and organs that transport lymph and immune cells. Lymphatic vessels collect lymph
from tissues, carry it to lymph nodes, and ultimately return it to the bloodstream. This
system facilitates the circulation of immune cells and helps drain excess fluids from
tissues.
​ Mucosa-Associated Lymphoid Tissue (MALT): MALT consists of immune
structures located in mucosal tissues, such as the gastrointestinal and respiratory
tracts. It includes structures like Peyer's patches in the small intestine and tonsils in
the throat. MALT plays a crucial role in immune defense at mucosal surfaces, where
the body is most exposed to pathogens.
​ Gut-Associated Lymphoid Tissue (GALT): GALT is a subset of MALT found in the
gastrointestinal tract. It includes specialized immune structures like Peyer's patches
and isolated lymphoid follicles. GALT is important for protecting against intestinal
infections and maintaining immune tolerance to dietary antigens.
​ Bronchus-Associated Lymphoid Tissue (BALT): BALT is a component of MALT
located in the respiratory tract, specifically the bronchi and lungs. It helps protect
against respiratory infections and contributes to local immune responses.
​ Skin-Associated Lymphoid Tissue (SALT): SALT includes immune structures
found in the skin, such as Langerhans cells and local immune cells. These cells are
part of the body's defense against skin infections and allergens.

2. Explain the patterns of clonal selection.


Clonal selection is a fundamental concept in immunology that describes how the immune
system generates a diverse and specific response to pathogens. It involves the activation
and proliferation of specific immune cells in response to the presence of antigens (molecules
that trigger an immune response). Here are the patterns of clonal selection:
​ Antigen Encounter: Clonal selection begins when the immune system encounters
an antigen, which can be a part of a pathogen, such as a protein on the surface of a
bacterium or a virus. Antigens are recognized by specific receptors on immune cells.
​ Antigen Recognition: Immune cells, particularly lymphocytes (B cells and T cells),
have antigen-specific receptors on their surfaces. When a lymphocyte's receptor
matches an antigen, it binds to the antigen, initiating the immune response.
​ Clonal Expansion: Upon antigen recognition, the specific immune cell undergoes
clonal expansion. This means that the activated immune cell rapidly divides to
produce a large number of identical daughter cells. These daughter cells are
collectively referred to as a "clone."
​ Diversity of Specificity: The immune system contains a vast repertoire of
lymphocytes, each with a unique antigen receptor. This diversity ensures that the
immune system can recognize and respond to a wide range of antigens. When an
antigen matches a specific receptor, only the lymphocytes with that particular
receptor are activated.
​ Effector Functions: The expanded clone of activated lymphocytes differentiates into
effector cells. B cells, for example, can become plasma cells that produce antibodies,
while T cells can differentiate into various types, such as cytotoxic T cells and helper
T cells.
​ Antigen Elimination: The effector cells generated from clonal expansion are now
equipped to eliminate the antigen. For instance, antibodies produced by plasma cells
can neutralize or tag pathogens, and cytotoxic T cells can kill infected host cells.
​ Memory Cells: Alongside the effector cells, some activated lymphocytes become
memory cells. These memory cells are long-lived and "remember" the specific
antigen. In future encounters with the same antigen, memory cells can mount a faster
and more effective response, leading to immunological memory.
​ Tolerance and Self-Antigens: Clonal selection is not only about responding to
pathogens. The immune system is also programmed to tolerate self-antigens,
preventing the activation of immune responses against the body's own tissues.
Autoimmune diseases occur when this tolerance mechanism fails.

3. Explain the importance of antigen-independent differentiation of lymphocytes.


Generation of Diverse Lymphocytes: Antigen-independent differentiation generates a
diverse pool of lymphocytes. During this phase, lymphocytes undergo genetic
rearrangements in their antigen receptor genes. In B cells, this involves the rearrangement
of immunoglobulin (antibody) genes, while in T cells, it's the T-cell receptor genes. This
genetic rearrangement results in a wide variety of antigen receptor specificities.
Recognition of a Broad Range of Antigens: The diversity of antigen receptor specificities
is crucial for the immune system to recognize and respond to a vast array of potential
antigens. Different pathogens present different antigens, and the immune system needs to
be prepared to target any possible threat. Antigen-independent differentiation ensures that
there are lymphocytes with receptors capable of recognizing a wide spectrum of antigens.
Self vs. Non-Self Discrimination: During antigen-independent differentiation, lymphocytes
also undergo a process of tolerance. This means that lymphocytes with receptors that
recognize self-antigens (antigens found in the body's own tissues) are either eliminated or
inactivated. This is critical for preventing autoimmune reactions where the immune system
mistakenly targets the body's own cells and tissues.
Maintenance of Immunological Memory: Some lymphocytes generated during
antigen-independent differentiation become memory cells. These memory cells "remember"
past encounters with antigens and can mount more rapid and efficient responses upon
re-exposure. This is the basis of immunological memory and the reason why the immune
system becomes more effective in protecting against previously encountered pathogens.
Versatility and Adaptability: The antigen-independent phase establishes the foundation for
the adaptive immune response. When lymphocytes encounter antigens that match their
specific receptors, they become activated and undergo clonal expansion, as explained by
clonal selection. This rapid response, based on pre-existing diversity, allows the immune
system to adapt to new infections quickly.
4. Explain the importance of antigen-dependent differentiation of lymphocytes.
​ Specificity and Memory Formation: Antigen-dependent differentiation allows
lymphocytes, such as B cells and T cells, to become activated in response to specific
antigens. When a lymphocyte encounters an antigen that matches its specific
receptor, it becomes activated, leading to the clonal selection and expansion of
lymphocytes with receptors that can effectively respond to that particular antigen.
This specificity is crucial because it ensures that the immune system can tailor its
response to the exact pathogen or antigen it encounters.
​ Generation of Effector Cells: During antigen-dependent differentiation, activated
lymphocytes differentiate into effector cells with specialized functions. For example, B
cells differentiate into plasma cells, which produce antibodies that can neutralize or
eliminate the antigen. T cells can differentiate into cytotoxic T cells, which are
responsible for killing infected or abnormal cells, or helper T cells, which coordinate
the immune response. This specialization allows the immune system to mount an
effective defense against different types of pathogens.
​ Immunological Memory: One of the key outcomes of antigen-dependent
differentiation is the generation of memory cells. Memory cells are long-lived
lymphocytes that "remember" the antigen they encountered during the initial immune
response. If the same antigen is encountered again in the future, memory cells can
rapidly initiate a more robust and quicker immune response. This is the basis of
immunological memory, which is responsible for the enhanced and more effective
protection against previously encountered pathogens.
​ Secondary Immune Response: Antigen-dependent differentiation is critical for the
secondary immune response. During a second encounter with the same antigen,
memory cells can be activated much faster than naïve (previously unexposed)
lymphocytes. This results in a more rapid and stronger immune response, often
preventing the pathogen from causing disease before it can establish an infection.
​ Adaptation to Varied Antigens: The adaptive immune system relies on
antigen-dependent differentiation to adapt to the wide variety of antigens that it may
encounter. This diversity of antigen receptors in the lymphocyte population allows the
immune system to recognize and respond to countless different pathogens, including
bacteria, viruses, fungi, and parasites. It also enables the immune system to target
specific variants of pathogens, making it effective against rapidly mutating viruses, for
example.
5. List the main types of lymphatic tissue cells and indicate their functions.
● B Cells: B cells are responsible for antibody production (humoral immunity). When
activated, they differentiate into plasma cells, which secrete antibodies that can
neutralize pathogens.
● T Cells: T cells play a central role in cell-mediated immunity. There are several
subsets of T cells, including helper T cells, cytotoxic T cells, and regulatory T cells,
each with specific functions.
● Helper T cells assist other immune cells in their functions, such as activating
B cells and cytotoxic T cells.
● Cytotoxic T cells directly attack and destroy infected or abnormal cells.
● Regulatory T cells help control the immune response and prevent it from
being overly aggressive or damaging healthy tissues.
6. Name and explain types of immune response.
​ Innate Immunity:
● Definition: Innate immunity is the body's first line of defense against
pathogens. It provides immediate, but non-specific protection.
● Characteristics:
● Present at birth and always active.
● Offers a rapid response to a wide range of pathogens.
● Lacks immunological memory, meaning it does not improve upon
repeated exposure to the same pathogen.
● Components of innate immunity include physical barriers (e.g., skin
and mucous membranes), cells (e.g., macrophages, neutrophils,
natural killer cells), and proteins (e.g., complement proteins).
● Functions:
● Physical barriers block the entry of pathogens.
● Phagocytic cells (e.g., macrophages and neutrophils) engulf and
destroy invading microorganisms.
● Natural killer (NK) cells target and kill infected or cancerous cells.
● Inflammatory responses are triggered to recruit immune cells to the
site of infection and facilitate pathogen clearance.
​ Adaptive (Acquired) Immunity:
● Definition: Adaptive immunity is a specific and highly tailored immune
response that develops throughout life in response to exposure to pathogens
or vaccines.
● Characteristics:
● Specificity: It targets specific pathogens or antigens.
● Memory: After an initial exposure, the immune system "remembers"
the pathogen, resulting in a faster and more effective response upon
re-exposure.
● Diversity: It can recognize a vast array of different antigens.
● Slower response time compared to innate immunity.
● Components:
● Lymphocytes, including B cells (responsible for antibody production)
and T cells (involved in cell-mediated immunity).
● Antibodies (immunoglobulins) produced by B cells.
● Major histocompatibility complex (MHC) molecules that help present
antigens to T cells.
● Functions:
● B cells produce antibodies (humoral immunity) that can neutralize
pathogens.
● T cells participate in cell-mediated immunity, helping destroy infected
or abnormal cells.
● The immune system generates memory cells (memory B and T cells)
that provide long-term protection by recognizing and responding
rapidly to previously encountered pathogens.
● Immunization, such as through vaccination, induces adaptive
immunity to specific pathogens without causing disease.

7. Discuss primary and secondary immune response.


The primary and secondary immune responses are stages in the adaptive immune system's
reaction to an antigen (e.g., a pathogen or a vaccine). They differ in terms of speed,
strength, and duration. Here's an explanation of both:
Primary Immune Response:
● First Encounter: When the immune system encounters a novel antigen for the first
time (e.g., during the first infection or vaccination), it initiates a primary immune
response.
● Lag Phase: The initial response is relatively slow, as it takes time for B cells and T
cells to recognize and respond to the antigen. This phase can last several days.
● Low Antibody Production: B cells differentiate into plasma cells, which produce
antibodies specific to the antigen. However, the production of antibodies is relatively
low during the primary response.
● IgM Predominance: In the early stages, IgM antibodies are the primary antibody
isotype produced. IgG production increases as the response progresses.
● Short Duration: The primary immune response may provide some level of protection,
but it is usually insufficient to completely eliminate the pathogen. It also takes time to
reach its peak.
● Memory Cell Formation: Memory B and T cells are generated, which "remember" the
antigen for future encounters.
Secondary Immune Response:
● Subsequent Encounters: If the same antigen is encountered again, the secondary
immune response is triggered. This can happen after re-infection with the same
pathogen or re-vaccination.
● Rapid and Strong: The secondary response is much faster and more robust. It
reaches its peak more quickly, often within hours.
● High Antibody Production: Memory B cells can rapidly differentiate into plasma cells,
leading to a higher production of specific antibodies. This results in higher antibody
titers.
● IgG Dominance: IgG antibodies predominate during the secondary response, and
they are more effective at neutralizing pathogens and providing protection.
● Long-Lasting: The secondary immune response provides more durable protection
due to the presence of memory cells. It can prevent the pathogen from causing
illness.
● Immunological Memory: Memory B and T cells respond rapidly and effectively to the
antigen. This is the basis of immunization, as it mimics the secondary response
without causing the disease.
In summary, the primary immune response occurs upon initial exposure to an antigen and is
characterized by a slower and less intense reaction. The secondary immune response
occurs upon re-exposure to the same antigen and is faster, stronger, and longer-lasting,
primarily due to the presence of memory cells. Immunization is based on the principle of
generating immunological memory to pathogens through the primary immune response, so
that the secondary response can provide more effective protection in case of future
encounters.

8. Describe and explain the interaction of immune cells in the immune response.
The immune response is a highly coordinated process involving various immune cells that
work together to recognize and eliminate pathogens. Here's an overview of how immune
cells interact during the immune response:
1. Antigen Presentation:
● The immune response begins when antigen-presenting cells (APCs), such as
dendritic cells, macrophages, and B cells, capture antigens from pathogens.
● APCs process these antigens and present fragments of them on their cell surface
using major histocompatibility complexes (MHC).
2. Activation of Helper T Cells:
● CD4+ T helper (Th) cells recognize the antigen-MHC complex on APCs and become
activated.
● Activated Th cells release cytokines, which are signaling molecules that instruct other
immune cells.
3. B Cell Activation:
● B cells recognize antigens directly or receive help from Th cells.
● Upon activation, B cells differentiate into plasma cells, which produce antibodies
specific to the pathogen.
● Memory B cells are also formed for long-term immunity.
4. Cytotoxic T Cell Activation:
● CD8+ cytotoxic T cells (CTLs) recognize infected cells displaying antigens on their
MHC class I molecules.
● CTLs are activated and become cytotoxic, capable of killing infected cells.
5. Macrophage Activation:
● Macrophages are further activated by Th cell cytokines to enhance their phagocytic
and killing abilities.
● They play a crucial role in engulfing and destroying pathogens.
6. Natural Killer (NK) Cells:
● NK cells are activated when they detect stressed or infected cells.
● They can directly kill target cells and secrete cytokines to enhance the immune
response.
7. Inflammatory Response:
● Neutrophils are rapidly recruited to sites of infection, where they engulf pathogens.
● The release of cytokines and chemokines leads to an inflammatory response,
recruiting immune cells to the infection site.
8. Immunological Memory:
● Memory B and T cells are formed during the response.
● In future encounters with the same pathogen, memory cells provide a quicker and
more robust response.
9. Describe the interaction between innate and adaptive immunity
Recognition of Pathogens:
● Innate Immunity: Innate immune cells, such as macrophages, neutrophils, and
dendritic cells, recognize and respond to common patterns associated with
pathogens. These patterns are known as pathogen-associated molecular patterns
(PAMPs).
● Adaptive Immunity: Adaptive immune cells, like T and B cells, recognize specific
antigens on pathogens. These antigens are presented to them by antigen-presenting
cells (APCs), mainly dendritic cells.
Activation of Innate Immunity:
● Innate Immunity: Upon detecting a pathogen, innate immune cells initiate an
immediate and non-specific response. They release cytokines and chemokines,
induce inflammation, and attempt to eliminate the threat through phagocytosis and
other mechanisms.
● Adaptive Immunity: The antigens captured and presented by APCs are recognized
by helper T cells (CD4+). These T cells receive signals from innate immune cells,
promoting their activation and differentiation into effector cells.
Coordinated Response:
● Innate Immunity: The innate immune response, with its rapid actions, serves as the
first line of defense. It helps contain the pathogen while adaptive immunity is gearing
up.
● Adaptive Immunity: Once activated, helper T cells release cytokines that guide B
cells and cytotoxic T cells. B cells produce pathogen-specific antibodies, and
cytotoxic T cells eliminate infected cells. These actions are highly specific and
tailored to the pathogen.
Feedback Mechanisms:
● Innate Immunity: Innate immune responses contribute to shaping the adaptive
immune response. Innate immune cells can provide co-stimulatory signals to T cells,
guiding their differentiation.
● Adaptive Immunity: Adaptive immune responses can modulate innate immune cells.
For instance, activated T cells can enhance the function of macrophages.
Memory Formation:
● Adaptive Immunity: One of the key features of adaptive immunity is the formation of
immunological memory. Memory B and T cells are generated, which provide
long-lasting protection upon re-exposure to the same pathogen. This is not a feature
of innate immunity.
Resolution of Immune Response:
● Regulatory T cells (Tregs) play a role in dampening immune responses to avoid
excessive inflammation and tissue damage. They help regulate both innate and
adaptive immune responses.
Overall, innate immunity serves as a rapid and non-specific defense mechanism that
provides immediate protection against a wide range of threats. Adaptive immunity, on the
other hand, is specific and tailored to the pathogen, providing long-lasting immunity through
memory cells. The interactions between these two branches ensure a comprehensive and
effective response to infections and other challenges while maintaining immune balance.
10. Explain the differences between innate and adaptive immunity.

11. Compare the functional characteristics of cells of innate and adaptive immunity.
Innate Immunity Cells:
​ Neutrophils:
● Phagocytosis of bacteria and debris.
● Release of antimicrobial molecules.
​ Macrophages:
● Phagocytosis of pathogens and dead cells.
● Antigen presentation to initiate adaptive immune response.
● Secretion of cytokines to coordinate immune responses.
​ Natural Killer (NK) Cells:
● Detection and destruction of infected or cancerous cells.
● Release of cytotoxic granules.
​ Dendritic Cells:
● Antigen presentation to T cells, initiating adaptive immunity.
● Cytokine production for immune regulation.
​ Basophils and Mast Cells:
● Release of inflammatory mediators like histamine.
● Role in allergic reactions.
Adaptive Immunity Cells:
​ T Cells (CD4+):
● Helper T cells assist B cells and cytotoxic T cells.
● Regulatory T cells control immune responses.
● Memory T cells provide long-term immunity.
​ T Cells (CD8+):
● Cytotoxic T cells directly kill infected or abnormal cells.
● Memory T cells respond rapidly upon re-exposure.
​ B Cells:
● Produce antibodies (immunoglobulins) targeting specific antigens.
● Memory B cells provide long-lasting antibody production.
​ Plasma Cells:
● Derived from B cells, produce large quantities of antibodies.
​ Antigen-Presenting Cells (APCs):
● Dendritic cells, macrophages, B cells present antigens to T cells.
● Initiate adaptive immune responses.
​ Memory Cells:
● Remember specific pathogens for faster response upon re-infection.
​ Cytokine-Producing Cells:
● Various immune cells produce cytokines to regulate immune responses.

Major histocompatibility complex and its general


organization.
1. Describe and explain the structure of the major histocompatibility complex.
The Major Histocompatibility Complex (MHC) is a critical component of the immune system,
responsible for presenting antigens to T cells, which play a central role in adaptive immunity.
There are two main classes of MHC molecules: MHC class I and MHC class II. Here's an
explanation of their structure and functions:
MHC Class I:
● Structure: MHC class I molecules are glycoproteins consisting of a heavy chain
(alpha chain) and a smaller subunit called beta-2 microglobulin. The heavy chain is
anchored in the cell membrane.
● Cellular Location: MHC class I molecules are found on the surface of virtually all
nucleated cells in the body.
● Function: Their primary role is to present endogenous antigens, such as viral or
intracellular bacterial proteins, to cytotoxic T cells (CD8+ T cells). This process helps
the immune system recognize and destroy infected or abnormal cells. It's essential
for immune surveillance against intracellular pathogens and tumor cells.
MHC Class II:
● Structure: MHC class II molecules consist of two glycoprotein chains (alpha and beta
chains) that are both anchored in the cell membrane.
● Cellular Location: MHC class II molecules are primarily found on the surface of
antigen-presenting cells (APCs) like dendritic cells, macrophages, and B cells.
● Function: MHC class II molecules present exogenous antigens, typically derived from
extracellular pathogens (e.g., bacteria), to helper T cells (CD4+ T cells). This
interaction is crucial for the activation of both cellular and humoral immune
responses. It allows T cells to recognize and respond to specific pathogens,
orchestrating the immune system's defense.
Both MHC class I and II molecules have a peptide-binding groove, which accommodates
short peptide fragments (antigens). The peptides bound by MHC molecules are derived from
proteins broken down within the cell (MHC class I) or from proteins taken up from the
extracellular environment (MHC class II).
MHC molecules are highly polymorphic, meaning they exist in numerous allelic forms within
a population. This diversity is essential for the immune system's ability to recognize a wide
range of antigens.
The interaction between MHC molecules and T cell receptors (TCRs) on T cells is a critical
step in antigen recognition and immune response. T cells recognize antigens when they are
presented by MHC molecules, enabling the immune system to launch targeted and specific
immune reactions against pathogens.
2. Describe the structure and function of the MHC I and MHC II molecules.
MHC I (Major Histocompatibility Complex Class I):
● Structure: MHC class I molecules are composed of a heavy chain (alpha chain) and
a smaller protein called beta-2 microglobulin. The heavy chain is anchored in the cell
membrane. Within the heavy chain, there are three domains: α1, α2, and α3. These
domains collectively form a peptide-binding groove.
● Cellular Location: MHC class I molecules are found on the surface of nearly all
nucleated cells in the body.
● Function: The primary role of MHC I molecules is to present endogenous antigens,
typically peptides derived from proteins synthesized within the cell. These antigens
are presented to cytotoxic T cells (CD8+ T cells). This process plays a crucial role in
immune surveillance, allowing the immune system to recognize and eliminate cells
infected with intracellular pathogens, such as viruses or intracellular bacteria, as well
as abnormal or cancerous cells.
MHC II (Major Histocompatibility Complex Class II):
● Structure: MHC class II molecules consist of two glycoprotein chains, an alpha chain
(α) and a beta chain (β), both anchored in the cell membrane. Each chain has two
domains (α1, α2, β1, and β2) that together create the antigen-binding site.
● Cellular Location: MHC class II molecules are primarily found on the surface of
antigen-presenting cells (APCs) such as dendritic cells, macrophages, and B cells.
● Function: MHC II molecules primarily present exogenous antigens, typically peptides
derived from proteins taken up from the extracellular environment (e.g., bacteria or
other pathogens), to helper T cells (CD4+ T cells). This interaction is crucial for
initiating both cellular and humoral immune responses. It allows T cells to recognize
specific pathogens and coordinate immune responses against those pathogens.
In both MHC I and MHC II, the peptide-binding groove accommodates short peptide
fragments, which are derived from protein breakdown processes within the cell (MHC I) or
from proteins captured from the extracellular environment (MHC II). The interaction between
MHC molecules and T cell receptors (TCRs) on T cells is central to antigen recognition and
the initiation of immune responses.
MHC molecules are highly polymorphic, with multiple allelic forms in the population, which
contributes to the immune system's ability to recognize a broad range of antigens.
3. Explain the principle of MHC inheritance
The principle of Major Histocompatibility Complex (MHC) inheritance refers to how MHC
genes, which code for MHC molecules, are passed from one generation to the next.
​ Codominant Inheritance: MHC genes are codominant, meaning that if an individual
has two different forms (alleles) of an MHC gene, both alleles are expressed. This is
in contrast to some other genes where one allele may dominate the expression of the
other (dominant-recessive inheritance). In the case of MHC, both alleles are active.
​ High Genetic Diversity: The MHC is one of the most polymorphic regions in the
human genome. This high polymorphism is maintained within populations because of
the advantage of having diverse MHC alleles. A diverse MHC allows an individual's
immune system to recognize a wide range of antigens, enhancing its ability to defend
against a variety of pathogens.
​ Inheritance from Each Parent: Humans inherit one MHC allele from each parent, so
they have two different MHC alleles (one from the mother and one from the father) at
each MHC gene locus. This contributes to the diversity of MHC molecules within a
population.
​ Recombination: Genetic recombination during meiosis can result in the creation of
new combinations of MHC alleles in offspring. This further adds to the genetic
diversity of MHC genes within a population.
​ Balancing Selection: Natural selection plays a role in maintaining the diversity of
MHC genes. Pathogens are constantly evolving, and the MHC diversity allows the
immune system to recognize and respond to new or changing pathogens effectively.
As a result, there is selective pressure to maintain MHC polymorphism.
In summary, the principle of MHC inheritance ensures that individuals inherit a diverse set of
MHC alleles from their parents, leading to a highly polymorphic MHC system that is vital for
the immune system's ability to recognize and combat a wide range of pathogens. This
diversity is maintained by the inheritance of two different MHC alleles, genetic
recombination, and the selective advantage of MHC polymorphism in the face of evolving
pathogens.

4. Explain the role of major histocompatibility complex gene products in the immune
Response.
he Major Histocompatibility Complex (MHC) gene products play a crucial role in the immune
response, as they are responsible for presenting antigens to immune cells. There are two
main classes of MHC gene products, MHC class I and MHC class II, each with distinct roles
in the immune system:
​ MHC Class I Molecules:
● Function: MHC class I molecules are found on the surface of nearly all
nucleated cells in the body. Their primary role is to present endogenous
antigens, which are typically fragments of proteins derived from within the
cell. These antigens can be viral proteins, tumor antigens, or other
intracellular proteins.
● Immune Response: When a cell is infected with a virus or becomes
cancerous, it will present viral or tumor antigens on its MHC class I
molecules. Cytotoxic T lymphocytes (CTLs or CD8+ T cells) survey these
MHC class I-antigen complexes. If the antigens are recognized as foreign or
abnormal, the CTLs will destroy the infected or cancerous cell to prevent the
infection from spreading.
​ MHC Class II Molecules:
● Function: MHC class II molecules are primarily found on antigen-presenting
cells (APCs), including dendritic cells, macrophages, and B cells. These
molecules are responsible for presenting exogenous antigens, which are
typically derived from outside the cell, such as those from bacteria, viruses, or
environmental sources.
● Immune Response: Antigen-presenting cells internalize and process
exogenous antigens. These antigens are then presented on their MHC class
II molecules. Helper T lymphocytes (CD4+ T cells) recognize these MHC
class II-antigen complexes. When helper T cells bind to these complexes,
they initiate and coordinate various immune responses. For instance, they
can activate B cells to produce antibodies and stimulate cytotoxic T cells to
kill infected cells.
In summary, MHC gene products are critical for the immune response as they facilitate the
recognition of antigens by T cells. MHC class I molecules present antigens from within
infected or cancerous cells, allowing cytotoxic T cells to eliminate the affected cells. MHC
class II molecules present antigens from external sources to helper T cells, which then
orchestrate the immune response by activating other immune cells. The interactions
between MHC molecules and T cells are central to adaptive immunity and the body's ability
to recognize and combat pathogens and abnormal cells.

5. Give examples of associations between HLA antigens and diseases.


Type 1 diabetes is an autoimmune disease in which the immune system mistakenly attacks
and destroys the insulin-producing beta cells in the pancreas. This results in a lack of insulin,
a hormone necessary for regulating blood sugar levels. The genetic component of type 1
diabetes is well-established, and specific HLA (Human Leukocyte Antigen) genes are
strongly associated with the disease.
In particular, HLA-DR (Human Leukocyte Antigen - DR) and HLA-DQ (Human Leukocyte
Antigen - DQ) alleles are involved in the genetic susceptibility to type 1 diabetes. Here's how
these HLA alleles are linked to the disease:
​ Antigen Presentation: HLA molecules are responsible for presenting fragments of
proteins (antigens) to immune cells, specifically T cells. These antigens are
recognized as "self" or "foreign" by the immune system.
​ Role of HLA-DR and HLA-DQ: HLA-DR and HLA-DQ alleles play a critical role in
antigen presentation. In individuals with type 1 diabetes, specific alleles of HLA-DR
and HLA-DQ are associated with the presentation of antigens related to the
insulin-producing beta cells.
​ Autoimmune Response: When immune cells, particularly T cells, encounter
antigens presented by HLA-DR and HLA-DQ molecules that are associated with beta
cell proteins, they may become activated. In individuals with a genetic predisposition
to type 1 diabetes, these activated T cells can target and attack beta cells, leading to
their destruction.
​ Insulin Deficiency: As the autoimmune response progresses and more beta cells
are destroyed, the ability to produce insulin diminishes. This results in inadequate
insulin levels in the body, leading to high blood sugar levels (hyperglycemia).
​ Onset of Type 1 Diabetes: Over time, as a significant number of beta cells are
damaged or destroyed, the symptoms of type 1 diabetes become evident. Individuals
may experience increased thirst, frequent urination, unexplained weight loss, and
other symptoms of high blood sugar.
It's important to emphasize that the HLA-DR and HLA-DQ alleles do not directly cause type
1 diabetes. Instead, they are genetic factors that increase susceptibility to the disease by
influencing the immune response to beta cell antigens. Environmental triggers, such as viral
infections, are also thought to play a role in initiating the autoimmune response in genetically
predisposed individuals.

Antigens
1.Define the terms: antigen, hapten, carrier.
Antigen: An antigen is a molecule or molecular structure that is recognized by the immune
system as a foreign invader or a threat. Antigens can trigger an immune response, including
the production of antibodies, to defend the body against infections. Antigens can be found on
the surface of pathogens like bacteria or viruses, as well as on the surface of cells,
allergens, or other foreign substances.

Hapten: A hapten is a small molecule that, on its own, does not induce an immune
response. However, when it binds to a larger carrier molecule, it can become immunogenic.
Haptens are typically too small to be recognized by the immune system directly, but when
they attach to a carrier molecule, they can elicit an immune response against the entire
complex, including the carrier.

Carrier: In the context of immunology, a carrier is a larger molecule that can bind to a hapten
and make it immunogenic. By attaching to the hapten, the carrier presents the combined
structure to the immune system, which can then recognize and mount an immune response
against the hapten-carrier complex. Carriers are often proteins or other macromolecules that
can effectively present the hapten to the immune system.

2. Describe the chemical properties of antigens.


Antigenicity: Antigens must possess the property of antigenicity, which means they can be
recognized by the immune system as foreign or non-self. This recognition is crucial for the
initiation of an immune response.

Immunogenicity: Not all antigens are equally immunogenic. Some antigens are highly
immunogenic and can elicit a strong immune response, while others may be weakly
immunogenic and require the help of adjuvants or other immune-stimulating molecules to
provoke a response.
Molecular Diversity: Antigens can be composed of a wide variety of molecules, including
proteins, carbohydrates, lipids, and nucleic acids. The diversity of these molecules allows
the immune system to recognize a broad range of potential threats.

Epitopes: Antigens often contain specific regions called epitopes or antigenic determinants.
Epitopes are the parts of the antigen that are recognized by antibodies or T-cell receptors.
These epitopes can be linear sequences of amino acids in proteins or specific arrangements
of other molecular structures.

Self-Antigens: While antigens are typically thought of as foreign molecules, the immune
system also recognizes self-antigens, which are molecules derived from the body's own
tissues. Normally, the immune system is tolerant of self-antigens to avoid attacking healthy
tissues, but autoimmune diseases can result from the loss of self-tolerance.

3. List the main features of antigens;


​ Antigenicity: Antigens must be capable of being recognized by the immune system
as foreign or non-self. This recognition is a fundamental characteristic of antigens.

​ Foreignness: Antigens are typically of non-self origin, meaning they are derived
from pathogens (e.g., bacteria, viruses, fungi, parasites) or foreign substances that
enter the body. The immune system distinguishes between self and non-self
antigens.

​ Molecular Diversity: Antigens can be composed of a wide range of molecular types,
including proteins, carbohydrates, lipids, nucleic acids, and even small chemicals.
This diversity allows the immune system to recognize a broad array of potential
threats.

​ Epitopes: Antigens contain specific regions called epitopes or antigenic
determinants. Epitopes are the portions of the antigen that are recognized by
antibodies or T-cell receptors. They can be linear sequences of amino acids in
proteins or specific configurations of other molecular structures.

​ Immunogenicity: The immunogenicity of an antigen refers to its ability to induce an
immune response. Some antigens are highly immunogenic and can provoke a strong
and specific immune response, while others may be weakly immunogenic and
require the use of adjuvants or other immune-stimulating substances.

​ Specificity: Antigens can be highly specific, meaning they can be recognized by
particular immune cells or antibodies that are tailored to them. This specificity
ensures that the immune response is directed against the specific pathogen or
substance.

​ Heterogeneity: Antigens may exist in various forms or variants. Pathogens, for
example, can have different strains or serotypes with distinct antigenic profiles. This
heterogeneity can impact the immune response and vaccine design.

​ Cross-Reactivity: Some antigens may share structural similarities with other
molecules, leading to cross-reactivity. This means that an immune response
generated against one antigen may also target a different antigen with a similar
structure. Cross-reactivity can have both beneficial and detrimental effects in immune
responses.

​ Antigen Processing and Presentation: Antigens are typically processed and
presented to immune cells by antigen-presenting cells (APCs) such as dendritic cells,
macrophages, and B cells. These APCs break down antigens into smaller fragments
and display them on their surfaces using molecules like major histocompatibility
complex (MHC) proteins. This presentation is essential for the activation of T cells
and the generation of an adaptive immune response.

​ Self-Antigens: While antigens are often thought of as foreign, the immune system
also recognizes self-antigens, which are molecules derived from the body's own
tissues. Normally, the immune system is tolerant of self-antigens to avoid attacking
healthy tissues, but autoimmune diseases can result from the loss of self-tolerance.

4. Explain role of antigenic determinants and carrier in the induction of immune


Response;
Antigenic determinants (also known as epitopes) and carriers play important roles in the
induction of immune responses, especially in the context of vaccines. Here's an explanation
of their roles:
​ Antigenic Determinants (Epitopes):
● Recognition: Antigenic determinants are specific regions or sites on the
surface of an antigen molecule that are recognized by immune cells,
particularly B cells and their antibodies, as well as T cells with specific
receptors. These epitopes are like molecular signatures that the immune
system uses to identify foreign substances.
● Immunogenicity: The immune system responds to the presence of antigenic
determinants. When the immune system encounters an antigen with epitopes
that match the receptors on immune cells (such as B cell receptors or T cell
receptors), it triggers an immune response.
● Diversity: Antigens can have multiple epitopes, each recognized by different
immune cells or antibodies. This diversity allows the immune system to
respond to a wide range of pathogens and foreign substances.
​ Carrier Proteins or Molecules:
● Enhanced Immunogenicity: In some cases, antigens themselves may not be
highly immunogenic, meaning they may not effectively trigger a strong
immune response when administered alone. Carrier proteins or molecules
can enhance the immunogenicity of these antigens.
● Conjugate Vaccines: Carrier molecules are often used in the development of
conjugate vaccines. These vaccines combine a less immunogenic antigen
(usually a carbohydrate antigen) with a carrier protein. The carrier protein,
which is typically highly immunogenic, helps the immune system recognize
and respond to the attached antigen more effectively.
● Example: Haemophilus influenzae type b (Hib) vaccine is a conjugate vaccine
that consists of a carbohydrate antigen (the capsular polysaccharide of Hib)
linked to a carrier protein. The carrier protein helps stimulate a robust immune
response against the carbohydrate antigen, providing protection against Hib
infections.
In summary, antigenic determinants (epitopes) are critical for the specific recognition of
antigens by the immune system, leading to the initiation of immune responses. Carrier
proteins or molecules can be used to enhance the immunogenicity of less potent antigens,
particularly in the development of vaccines, by providing a strong immune-stimulating
component that aids in the recognition and response to the attached antigen. This approach
is important for vaccine development, especially when dealing with antigens that are
otherwise weak inducers of immune responses.

5. Describe the thymus-dependent and thymus-independent antigens.


Thymus-dependent (TD) and thymus-independent (TI) antigens are two categories of
antigens that differ in their ability to stimulate an immune response and their dependence on
the presence of the thymus and T cells. These distinctions are important in understanding
the nature of immune responses to various antigens.

Thymus-Dependent (TD) Antigens:


​ Definition: TD antigens are antigens that require the assistance of T helper (Th) cells,
a type of T lymphocyte, to induce a robust and effective immune response. They are
often protein-based antigens.
​ Processing: When TD antigens are encountered by antigen-presenting cells (APCs)
like dendritic cells or macrophages, these cells process the antigens and present
peptide fragments on their surfaces using major histocompatibility complex class II
(MHC-II) molecules.
​ T Cell Activation: Th cells recognize the antigenic peptides presented on APCs
through their T cell receptors (TCRs). This interaction activates Th cells, which, in
turn, provide signals and cytokines to B cells to stimulate their activation and
differentiation into plasma cells. Plasma cells produce antibodies specific to the TD
antigen.
​ Immunological Memory: TD antigen responses typically lead to the development of
immunological memory, meaning that the immune system "remembers" the antigen,
allowing for a faster and more robust response upon re-exposure to the same
antigen.
​ Examples: Many bacterial and viral proteins are TD antigens. Common vaccines, like
those for tetanus and diphtheria, target TD antigens.

Thymus-Independent (TI) Antigens:
​ Definition: TI antigens are antigens that can initiate an immune response without the
direct involvement of T helper cells. They are often polysaccharides or molecules
with repeating, highly conserved structures.
​ Limited T Cell Involvement: TI antigens can activate B cells directly, as B cells have
receptors (B cell receptors or BCRs) capable of recognizing certain TI antigens.
However, T cells may still play a supporting role in TI antigen responses.
​ Poor Immunological Memory: TI antigen responses typically do not generate strong
immunological memory, so repeat exposures may not result in a more robust
response.
​ Examples: Bacterial polysaccharides, such as those found in the capsules of certain
bacteria like Streptococcus pneumoniae, are TI antigens. Vaccines against these
pathogens often incorporate conjugate vaccines, which combine TI antigens with
carrier proteins to enhance their immunogenicity and generate a more effective
response.
In summary, the distinction between thymus-dependent (TD) and thymus-independent (TI)
antigens lies in their dependence on T helper cells, the nature of their recognition by B cells,
and the resulting immune responses. TD antigens require T helper cells for a robust immune
response, while TI antigens can initiate responses without direct T cell involvement, although
T cells may still play some role. Understanding these differences is crucial for vaccine
development and immunological research.

6. Explain mechanisms of formation of the immune response to thymus-dependent


and thymus-independent antigens.
The mechanisms of immune responses to thymus-dependent (TD) and thymus-independent
(TI) antigens differ based on the involvement of T cells and B cells. Here's an overview of
how the immune response is formed for each type of antigen:

Thymus-Dependent (TD) Antigens:

​ Antigen Recognition:
● TD antigens are typically protein-based antigens. When antigen-presenting
cells (APCs), such as dendritic cells or macrophages, encounter TD antigens,
they phagocytose the antigens and process them into smaller peptide
fragments.
● These peptide fragments are presented on the surface of APCs using major
histocompatibility complex class II (MHC-II) molecules.
​ T Cell Activation:
● T helper (Th) cells, a subset of T lymphocytes, recognize the antigenic
peptides presented by APCs through their T cell receptors (TCRs).
● This interaction activates Th cells, leading to their proliferation and
differentiation into effector Th cells and memory Th cells.
​ B Cell Activation:
● Activated Th cells provide help to B cells by releasing cytokines and cell
surface interactions.
● B cells have surface receptors called B cell receptors (BCRs) that can
recognize specific antigenic epitopes.
● When BCRs on B cells bind to the TD antigen, it initiates B cell activation.
​ B Cell Differentiation:
● Activated B cells undergo clonal expansion and differentiation into plasma
cells.
● Plasma cells produce large quantities of antibodies (immunoglobulins)
specific to the TD antigen.
​ Immune Response:
● Antibodies produced by plasma cells circulate in the bloodstream and can
neutralize or mark the TD antigen for destruction by other immune cells.
● Memory B cells are also generated during this process, providing long-term
immunity upon re-exposure to the same antigen.

Thymus-Independent (TI) Antigens:

​ Antigen Recognition:
● TI antigens are often polysaccharides, molecules with repeating structures, or
other highly conserved structures.
● B cells can directly recognize and bind to TI antigens using their BCRs
without the need for T cell assistance.
​ B Cell Activation:
● Binding of TI antigens to BCRs triggers signaling pathways within B cells,
leading to their activation.
​ Antibody Production:
● Activated B cells differentiate into plasma cells and produce antibodies
specific to the TI antigen.
​ Immune Response:
● Antibodies produced in response to TI antigens can neutralize the antigen or
opsonize it, making it more susceptible to phagocytosis by macrophages.
● TI antigen responses often generate weaker and shorter-lived immune
memory compared to TD antigen responses.
In summary, the key distinction between TD and TI antigens lies in the requirement for T cell
help in the immune response. TD antigens require T cell assistance to stimulate B cells
effectively, resulting in stronger and more durable immune responses, including the
formation of memory B cells. TI antigens, on the other hand, can directly activate B cells but
typically result in weaker and less durable immune responses with limited memory cell
formation.

7. Compare and contrast thymus-dependent and thymus- independent antigens.


Summary of Comparison:
● Nature: TD antigens are protein-based, while TI antigens are often non-protein
antigens.
● T Cell Involvement: TD antigens require T cell help, while TI antigens do not depend
on T cells for activation.
● Processing: TD antigens are processed by APCs and presented using MHC-II, while
TI antigens are recognized directly by BCRs.
● Immune Response: TD antigen responses are stronger, specific, and produce
memory B cells, while TI antigen responses are weaker and generate limited
memory.
● Vaccine Types: Many traditional vaccines are based on TD antigens, while some
pathogens with TI antigens require conjugate vaccines to enhance their
immunogenicity.
In summary, the key distinction between TD and TI antigens is their dependence on T cell
help, which significantly influences the strength and nature of the immune response elicited
against them.

8. Contrast primary and secondary self-antigens. Give examples of each.


Primary self-antigens and secondary self-antigens are two categories of self-antigens that
differ in their recognition by the immune system and their role in autoimmune responses.
Here's a contrast between these two types of self-antigens with examples:

Primary Self-Antigens:
​ Nature of Antigens:
● Primary self-antigens are normal, healthy self-antigens that are part of the
body's own tissues and organs.
​ Recognition:
● The immune system is generally tolerant of primary self-antigens and does
not mount an immune response against them.
● In a healthy individual, primary self-antigens do not typically trigger
autoimmune reactions.
​ Examples:
● Proteins, carbohydrates, lipids, and other molecules present in healthy cells
and tissues are considered primary self-antigens.
● Examples include the proteins in muscle cells, carbohydrates on the surface
of red blood cells, and self-antigens found in the skin.
​ Tolerance:
● The immune system is tolerant to primary self-antigens due to mechanisms
like central tolerance in the thymus and bone marrow, which eliminate or
inactivate autoreactive immune cells during development.

Secondary Self-Antigens:

​Nature of Antigens:
● Secondary self-antigens are self-antigens that become targets of an
autoimmune response due to a breakdown in self-tolerance mechanisms.
​ Recognition:
● Unlike primary self-antigens, the immune system recognizes secondary
self-antigens as foreign or non-self, leading to autoimmune reactions.
● Secondary self-antigens are associated with autoimmune diseases.
​ Examples:
● Secondary self-antigens include self-antigens that have been modified or
presented in an abnormal context, making the immune system perceive them
as threats.
● For example, in autoimmune diseases like rheumatoid arthritis, joint tissues
contain secondary self-antigens that are recognized as foreign, triggering an
autoimmune attack on the joints.
● In type 1 diabetes, beta cells in the pancreas are targeted by the immune
system as secondary self-antigens, resulting in the destruction of these
insulin-producing cells.
​ Breakdown of Tolerance:
● Autoimmune diseases often result from a breakdown in the mechanisms that
maintain self-tolerance. This breakdown can occur due to genetic
predisposition, environmental factors, or infections that expose the immune
system to self-antigens in an abnormal way.
Summary of Contrast:
● Nature: Primary self-antigens are normal, healthy self-antigens, while secondary
self-antigens are self-antigens that trigger autoimmune responses.
● Recognition: Primary self-antigens are tolerated by the immune system, while
secondary self-antigens are recognized as foreign due to a loss of self-tolerance.
● Examples: Primary self-antigens include those found in healthy tissues, while
secondary self-antigens are associated with autoimmune diseases and result from
abnormal immune responses to self-components.
● Tolerance Mechanisms: Primary self-antigens are tolerated due to effective central
and peripheral tolerance mechanisms, while autoimmune diseases often result from
the breakdown of these tolerance mechanisms, leading to the recognition of
secondary self-antigens as non-self.

Humoral factors of Immunity B-cell maturation.


1. Describe development of B-lymphocytes.

The development of B-lymphocytes, also known as B cells, is a complex process that occurs
primarily in the bone marrow. B cells are a critical component of the adaptive immune
system and play a central role in producing antibodies to fight infections. Here is an overview
of the development of B-lymphocytes:
​ Origins in Hematopoietic Stem Cells (HSCs): B cell development begins with
hematopoietic stem cells (HSCs) in the bone marrow. These stem cells have the
potential to give rise to all blood cells, including B cells.
​ Commitment to B Cell Lineage: Under the influence of specific signaling molecules
and transcription factors, some HSCs commit to the B cell lineage. This commitment
involves the activation of genes that specify the B cell developmental program.
​ Progenitor Cells: Committed HSCs differentiate into B cell progenitor cells, known
as pro-B cells. At this stage, specific genetic rearrangements occur in the DNA,
leading to the formation of a unique B cell receptor (BCR) on the cell's surface. The
BCR is a membrane-bound antibody that will allow the B cell to recognize specific
antigens.
​ Rearrangement of BCR Genes: During the pro-B cell stage, genes encoding the
variable regions of the BCR, which determine its antigen-binding specificity, undergo
a process called V(D)J recombination. This genetic rearrangement involves the
random joining of different gene segments (variable, diversity, and joining segments)
to generate a diverse repertoire of BCRs. This diversity is crucial for the ability of B
cells to recognize a wide range of antigens.
​ Checkpoint and Selection: Pro-B cells that successfully rearrange their BCR genes
undergo a selection process. Those cells with BCRs that bind to self-antigens too
strongly are eliminated (negative selection) to prevent autoimmune reactions. B cells
with functional BCRs that recognize antigens at an appropriate level proceed to the
next stage.
​ Maturation and Differentiation: The selected B cells continue to mature and
differentiate as they progress through various developmental stages, including pre-B
cells and immature B cells. These cells are tested for self-reactivity and further
refined.
​ Exit to the Periphery: Once B cells have completed their maturation and their BCRs
are properly assembled, they exit the bone marrow and enter the peripheral lymphoid
tissues, such as the spleen and lymph nodes.
​ Antigen Encounter and Activation: In peripheral lymphoid tissues, B cells circulate
through the bloodstream and lymphatic system. When a mature B cell encounters its
cognate antigen (the specific antigen it can bind to), it becomes activated. This
activation may occur when the BCR interacts with an antigen, often with the help of T
cells.
​ Proliferation and Differentiation: Upon activation, B cells undergo clonal
expansion, leading to the production of a large number of identical B cells. These
activated B cells can differentiate into plasma cells, which are antibody-producing
factories, or memory B cells that provide long-term immunity.
​ Antibody Production: Plasma cells are highly specialized and secrete large
quantities of antibodies into the bloodstream. These antibodies recognize and
neutralize the specific antigen encountered by the B cell.

2. Explain the ability of B lymphocytes to the immune response in the process of


Differentiation.
3. Explain how B cells recognize and respond to an antigen

​ Antigen Encounter: B cells are equipped with B cell receptors (BCRs) on their cell
surfaces. Each B cell has a unique BCR with a specific binding site. When an
antigen, such as a pathogen's protein or other foreign molecule, enters the body, it
interacts with the BCR of a B cell if the BCR matches the antigen's shape.
​ BCR-Antigen Binding: If the BCR on a B cell's surface matches the antigen's
shape, the antigen binds to the BCR, creating an antigen-BCR complex. This binding
is highly specific, similar to a lock and key mechanism. Only B cells with BCRs that
can recognize a particular antigen will respond.
​ Activation: The binding of the antigen to the BCR is the trigger for the B cell's
activation. It sets off a cascade of intracellular events, including signaling pathways,
leading to the B cell's activation.
​ Internalization and Antigen Processing: Once the antigen is bound to the BCR,
the B cell internalizes the antigen-BCR complex. Inside the B cell, the antigen is
processed and broken down into smaller fragments.
​ Antigen Presentation: B cells, like other antigen-presenting cells (APCs), present
the processed antigen fragments on their cell surface using a protein complex called
major histocompatibility complex II (MHC II). MHC II presents the antigen to other
immune cells, such as helper T cells (CD4+ T cells).
​ Helper T Cell Activation: Helper T cells recognize the antigen-MHC II complex
presented by B cells. When a helper T cell's T cell receptor (TCR) matches the
presented antigen, it becomes activated. The activation of helper T cells triggers a
complex immune response.
​ Co-Stimulation: Helper T cells require additional signals from the B cell for full
activation. This co-stimulation is usually provided by molecules known as
co-stimulatory proteins, such as CD40 on the B cell interacting with CD40L on the
helper T cell.
​ Clonal Expansion: Once fully activated, B cells undergo clonal expansion, where
they rapidly divide and proliferate. This results in a population of B cells, all derived
from the original antigen-specific B cell.
​ Differentiation: Some of the newly generated B cells differentiate into plasma cells.
Plasma cells are specialized antibody factories that produce and secrete large
quantities of antibodies specific to the antigen. These antibodies can neutralize the
pathogen and tag it for destruction by other immune cells.
​ Memory B Cells: In addition to plasma cells, some B cells differentiate into memory
B cells. These cells have a long lifespan and "remember" the specific antigen.
Memory B cells provide long-term immunity, ensuring a faster and stronger immune
response if the same antigen is encountered again in the future.
4. List the markers of B-cells.

5. Describe B-cell receptor.

6. Summarize role of humoral immunity in immune reactions.

Development of B-lymphocytes
1. Development of B-lymphocytes.
B-lymphocytes, or B cells, are a type of white blood cell that plays a vital role in the immune
system. The development of B-lymphocytes occurs primarily in the bone marrow and
involves several stages of differentiation and maturation. Here is an overview of the
development of B-lymphocytes:
​ Hematopoietic Stem Cells (HSCs): B-cell development begins with hematopoietic
stem cells (HSCs) in the bone marrow. These stem cells have the potential to
differentiate into various blood cell types, including B cells.
​ Early Progenitor Stage: HSCs commit to becoming lymphoid progenitor cells, which
are early precursors of both B cells and T cells. These progenitor cells migrate to
specific areas within the bone marrow known as lymphoid niches.
​ Expression of B-Cell Receptor (BCR): In the lymphoid niches of the bone marrow,
lymphoid progenitor cells start expressing a B-cell receptor (BCR) on their cell
surface. The BCR is a membrane-bound antibody (immunoglobulin) molecule
specific to a particular antigen.
​ Rearrangement of BCR Genes: One of the key events in B-cell development is the
rearrangement of the genes that encode the variable regions of the BCR. This
process is called V(D)J recombination and results in the generation of a diverse
repertoire of B cells, each capable of recognizing a unique antigen.
​ Expression of Pre-B Cell Receptor: Once the V(D)J recombination process is
successful and the BCR is expressed, the cells are referred to as pre-B cells. Pre-B
cells also express a surrogate light chain (SLC) to form a pre-B cell receptor
complex. This complex serves as a checkpoint to ensure that the rearranged BCR is
functional.
​ Positive Selection: Pre-B cells that have functional BCRs undergo positive
selection, allowing them to survive and proceed with their development.
Non-functional pre-B cells are eliminated.
​ Maturation and Migration: After positive selection, the pre-B cells continue to
mature and undergo further differentiation, eventually giving rise to immature B cells.
These immature B cells leave the bone marrow and migrate to secondary lymphoid
organs, such as the spleen and lymph nodes.
​ Encounter with Antigens: In secondary lymphoid organs, immature B cells
encounter antigens. If an immature B cell's BCR matches an antigen, it gets activated
and differentiates into either plasma cells or memory B cells.
​ Plasma Cells and Memory B Cells: Plasma cells are responsible for producing
antibodies specific to the encountered antigen, while memory B cells "remember" the
antigen for a faster immune response upon re-exposure.
In summary, the development of B-lymphocytes involves a series of stages in the bone
marrow, beginning with hematopoietic stem cells and culminating in the generation of mature
B cells capable of recognizing and responding to specific antigens. These B cells play a
crucial role in the adaptive immune response by producing antibodies and forming
immunological memory.
2. B lymphocytes differentiation.
The differentiation of B lymphocytes, or B cells, is a complex process that occurs primarily in
the bone marrow and results in the generation of mature B cells capable of producing
antibodies. Here is an overview of the differentiation of B cells:
​ Hematopoietic Stem Cells (HSCs): B-cell development begins with hematopoietic
stem cells (HSCs) in the bone marrow. These multipotent stem cells have the
potential to differentiate into various blood cell types, including B cells.
​ Commitment to the Lymphoid Lineage: HSCs commit to becoming lymphoid
progenitor cells, which are early precursors of both B cells and T cells. This
commitment is influenced by specific transcription factors.
​ Development of Pro-B Cells:
● Lymphoid progenitor cells progress to the pro-B cell stage. At this stage, they
express surface markers such as CD19, CD20, and CD22.
● One of the hallmark events in B-cell differentiation is the rearrangement of
immunoglobulin heavy chain (IgH) gene segments. This process is called
V(D)J recombination and leads to the formation of a unique B-cell receptor
(BCR).
​ Expression of Pre-BCR:
● Once the V(D)J recombination of the IgH gene is successful and a functional
heavy chain is produced, the pro-B cell expresses a pre-B cell receptor
(pre-BCR). The pre-BCR consists of the newly formed heavy chain and a
surrogate light chain (SLC).
● The pre-BCR plays a critical role in signaling and development, ensuring that
the BCR is functional.
​ Positive Selection:
● Pro-B cells with a functional pre-BCR undergo positive selection, leading to
their survival and progression to the next stage.
● Positive selection checks for the functionality of the pre-BCR and its ability to
signal the cell.
​ Development of Immature B Cells:
● After positive selection, the pro-B cells mature into pre-B cells and then into
immature B cells.
● Immature B cells express both IgM and IgD BCR isotypes on their surface.
​ Tolerance Checkpoints:
● Immature B cells undergo tolerance checkpoints in the bone marrow to
ensure that they do not react strongly to self-antigens. B cells with strongly
self-reactive BCRs are either eliminated or undergo receptor editing to modify
their specificity.
​ Migration to Secondary Lymphoid Organs:
● Mature, non-self-reactive B cells leave the bone marrow and migrate to
secondary lymphoid organs, such as the spleen and lymph nodes, where they
encounter antigens.
​ Encounter with Antigens:
● In secondary lymphoid organs, mature B cells encounter antigens. If the BCR
of a mature B cell matches an antigen, it gets activated.
​ Differentiation into Effector B Cells:
● Activated B cells differentiate into effector B cells, including plasma cells and
memory B cells.
● Plasma cells are responsible for producing antibodies specific to the
encountered antigen.
● Memory B cells "remember" the antigen and provide long-term immunological
memory for future encounters.
In summary, the differentiation of B lymphocytes involves multiple stages, from
hematopoietic stem cells to the generation of mature B cells. This process includes genetic
rearrangements, positive selection, tolerance checkpoints, and antigen encounter, ultimately
leading to the production of antibodies and immunological memory.

3. Markers of B-cells.
B cells, also known as B lymphocytes, are immune cells that play a central role in the
adaptive immune response. They can be identified by the presence of specific cell surface
markers or antigens. These markers are often used to characterize and distinguish B cells
from other immune cells. Some of the key markers or antigens associated with B cells
include:
​ CD19: CD19 is a transmembrane glycoprotein that is considered one of the most
reliable markers for identifying B cells. It is expressed throughout B-cell development,
from the pro-B cell stage to mature B cells. CD19 is a crucial component of the B-cell
receptor (BCR) complex and plays a role in B-cell signaling.
​ CD20: CD20 is another cell surface marker expressed on B cells. It is present on
pre-B cells and mature B cells, but not on plasma cells. CD20 is often targeted in the
treatment of B-cell malignancies, such as non-Hodgkin lymphoma, using monoclonal
antibody therapies like rituximab.
​ CD21 (Complement Receptor 2, CR2): CD21 is a receptor for complement
fragments, and it is expressed on mature B cells. It plays a role in B-cell activation
and antigen presentation.
​ CD22: CD22 is a B-cell-specific adhesion molecule and inhibitory receptor. It helps
regulate B-cell activation by modulating BCR signaling. CD22 is expressed on the
surface of mature B cells.
​ CD24: CD24 is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein
found on B cells. It is also expressed on other immune cells, but it can be used as a
marker for B-cell populations in combination with other markers.
​ IgM (Immunoglobulin M): IgM is the first immunoglobulin isotype expressed on the
surface of B cells as part of the BCR. It is associated with early B-cell development
and is present on naive B cells.
​ IgD (Immunoglobulin D): IgD is another immunoglobulin isotype that, like IgM, is
expressed on the surface of mature B cells as part of the BCR. It is involved in
antigen recognition.
​ HLA-DR (Human Leukocyte Antigen - DR isotype): HLA-DR is a major
histocompatibility complex (MHC) class II molecule that is upregulated on the surface
of B cells when they are activated. It plays a role in antigen presentation to helper T
cells.
​ CD38: CD38 is expressed on B cells, particularly plasma cells. It is involved in
calcium signaling and can serve as a marker for plasma cell populations.
​ CD138 (Syndecan-1): CD138 is a marker for plasma cells and is not typically found
on the surface of B cells. It is commonly used to identify and characterize plasma
cells in tissues.
These markers are useful for immunophenotyping and studying different stages of B-cell
development, activation, and function. Additionally, they are valuable in clinical diagnostics
and research related to B-cell disorders and immune responses

4. B-cell receptor.
The B-cell receptor (BCR) is a crucial component of B lymphocytes (B cells) and plays a
central role in the adaptive immune response. It is a membrane-bound protein complex that
allows B cells to recognize and bind to specific antigens, which are typically parts of
pathogens such as bacteria, viruses, or other foreign invaders. The BCR consists of several
key elements:
​ Immunoglobulin (Ig) M or IgD: The core of the BCR is an immunoglobulin
molecule, either IgM or IgD, which serves as the antigen-binding component. Each B
cell expresses surface IgM and IgD molecules with identical antigen-binding regions.
These immunoglobulins are specific to particular antigens.
​ Membrane-Bound Antibodies: IgM and IgD antibodies are anchored to the B-cell
membrane, with their antigen-binding regions exposed on the cell surface. These
antibodies are identical to the soluble antibodies (immunoglobulins) secreted by
plasma cells that develop from activated B cells.
​ Igα and Igβ: These are accessory proteins that are associated with the
immunoglobulin molecules in the BCR complex. Igα and Igβ play a crucial role in
signal transduction. When the BCR binds to an antigen, it induces phosphorylation of
these accessory proteins, initiating intracellular signaling cascades.
5. Role of humoral immunity in immune reactions.
Humoral immunity is a critical component of the adaptive immune system responsible for
defending the body against pathogens, particularly bacteria, viruses, and toxins. It involves
the production of antibodies (also known as immunoglobulins) by B lymphocytes (B cells)
and plays several essential roles in immune reactions:
​ Antibody Production: B cells, activated by exposure to specific antigens,
differentiate into plasma cells. Plasma cells are specialized antibody factories that
produce large quantities of antibodies with precise antigen specificity. Each B cell
produces antibodies with a unique antigen-binding site.
​ Antigen Neutralization: Antibodies produced by plasma cells can directly bind to
antigens, such as proteins on the surface of bacteria or viruses. This binding process
neutralizes the infectivity of pathogens by preventing them from attaching to and
entering host cells. In the case of viruses, neutralization can also prevent viral
replication.
​ Opsonization: Antibodies can coat pathogens, a process known as opsonization.
This makes the pathogens more recognizable to phagocytic cells, such as
macrophages and neutrophils. Phagocytes can then engulf and digest the opsonized
pathogens more efficiently, enhancing the body's defense against infections.
​ Complement Activation: Antibodies can trigger the complement system, a group of
blood proteins that enhance the immune response. Activation of the complement
system leads to several immune functions, including the formation of membrane
attack complexes (MACs) that can lyse bacterial cells and the recruitment of
inflammatory cells to the site of infection.
​ Antibody-Dependent Cellular Cytotoxicity (ADCC): Antibodies can facilitate the
destruction of infected or abnormal cells. Natural killer (NK) cells and certain types of
white blood cells, like macrophages, have Fc receptors that can bind to the Fc region
of antibodies. When antibodies bind to the surface of target cells (e.g., virus-infected
cells or cancer cells), these effector cells can recognize and kill the target cells.
​ Memory Response: After the initial encounter with an antigen, some B cells
differentiate into memory B cells. Memory B cells "remember" the specific antigen,
and upon re-exposure to the same antigen, they can rapidly generate a robust
antibody response. This is the basis for immunological memory and long-term
protection against pathogens.
​ Variability: Humoral immunity generates a diverse repertoire of antibodies, each
specific to a particular antigen. This diversity allows the immune system to respond
effectively to a wide range of pathogens and adapt to new challenges.
​ Duration of Protection: Humoral immunity provides both immediate and long-lasting
protection. While antibodies are rapidly produced during an active infection, memory
B cells ensure that the immu jim ne system can respond more quickly and effectively
to subsequent exposures to the same pathogen.
In summary, humoral immunity, mediated by antibodies produced by B cells, is a crucial arm
of the immune system that targets extracellular pathogens, toxins, and abnormal cells. It
plays a pivotal role in preventing and controlling infections, providing both immediate
defense and long-term immunological memory.
6. Antigen-dependent differentiation of B-lymphocytes.
Antigen-dependent differentiation of B-lymphocytes is a crucial process in the adaptive
immune response, leading to the development of plasma cells that produce antibodies
against specific antigens. This process involves several stages:
​ Antigen Encounter: The activation of B-lymphocytes begins when they encounter
an antigen that matches the antigen receptors on their surface. B cell receptors
(BCRs), which are membrane-bound antibodies, play a central role in antigen
recognition. These BCRs are immunoglobulin molecules anchored to the B cell
membrane.
​ Antigen Recognition: When an antigen binds to the BCRs on the B cell's surface, it
triggers a series of intracellular signaling events. This antigen recognition is highly
specific, with each B cell typically recognizing a single antigen or a closely related
group of antigens.
​ Internalization and Antigen Processing: After binding to the antigen, the B cell
internalizes the antigen-bound BCR complex through receptor-mediated endocytosis.
Once inside the B cell, the antigen is processed into smaller peptide fragments.
These antigen fragments are then displayed on the B cell's surface bound to major
histocompatibility complex class II (MHC-II) molecules.
​ Antigen Presentation: B cells, like other antigen-presenting cells (APCs), can
present the processed antigen fragments to helper T-lymphocytes (T cells) bearing
CD4 receptors. This interaction occurs within secondary lymphoid organs, such as
lymph nodes and the spleen. T cells are crucial in providing additional activation
signals to B cells.
​ T-Cell Help: When a helper T cell recognizes the antigen-MHC-II complex presented
by the B cell, it becomes activated. The activated helper T cell releases cytokines,
particularly interleukin-4 (IL-4) and interleukin-21 (IL-21), which play essential roles in
B-cell activation and differentiation.
​ B-Cell Activation: With the help of cytokines from activated helper T cells, the B cell
receives the necessary signals to become fully activated. This activation leads to
B-cell proliferation and differentiation.
​ Differentiation into Plasma Cells: Activated B cells differentiate into plasma cells,
also known as effector B cells. Plasma cells are specialized antibody factories that
produce large quantities of antibodies with the same antigen specificity as the original
B cell receptor. Each plasma cell produces a single type of antibody.
​ Antibody Production: The newly formed plasma cells release antibodies
(immunoglobulins) into the bloodstream. These antibodies circulate throughout the
body, binding to the specific antigen for which they were activated. Antibodies play
essential roles in immune defense, such as neutralizing pathogens, opsonizing them
for phagocytosis, activating the complement system, and facilitating
antibody-dependent cellular cytotoxicity (ADCC).
​ Memory B Cells: Alongside the generation of plasma cells, a subset of activated B
cells differentiates into memory B cells. Memory B cells "remember" the antigen and
remain in the body for a prolonged period. If the same antigen is encountered in the
future, memory B cells can quickly differentiate into plasma cells, resulting in a rapid
and robust antibody response.
Antigen-dependent differentiation of B-lymphocytes is a highly coordinated and specific
process that ensures the production of antibodies tailored to the encountered pathogen. This
process is a fundamental mechanism of adaptive immunity, allowing the immune system to
mount specific and memory responses to a wide range of antigens.

Structure and functions of antibodies.


1. Describe the structure and five types of antibodies.
Antibodies, also known as immunoglobulins (Ig), are essential proteins of the immune
system that play a crucial role in defending the body against infections. Antibodies have a
characteristic structure and are classified into several types or classes. Here's an overview
of antibody structure and five major types of antibodies:

Antibody Structure Notes:


● Light chains: Consist of one V (variable) and one C (constant) domain.
● Heavy chains: Consist of one V and three or four C domains.
● Each domain has a characteristic three-dimensional shape known as the
immunoglobulin (Ig) domain.
Antigen-Binding Site:
● Composed of the V regions of both heavy and light chains.
● Core antibody structure has two identical antigen-binding sites.
● Each variable region (VH or VL) contains three hypervariable regions called CDRs
(complementarity-determining regions).
● CDR3, at the VH and VL junction, exhibits the greatest variability.
Fab Fragment:
● Binds to antigens.
● Composed of one constant and one variable domain of both heavy and light chains.
● Shapes the paratope at the amino terminal end of the monomer.
● Two variable domains bind to specific antigen epitopes.
Fc Region:
● Located at the base of the Y-shaped antibody.
● Composed of two heavy chains with two or three constant domains, depending on
the antibody class.
● Regulates immune cell activity and mediates various physiological effects:
● Binds to specific proteins to ensure an appropriate immune response.
● Interacts with cell receptors, like Fc receptors, and immune molecules, such
as complement proteins.
● Mediates effects like opsonization, cell lysis, and degranulation of immune
cells.
IgG Molecule:
● Y-shaped structure with two light chains and two heavy chains.
● Each light chain contains variable and constant regions.
● Each heavy chain consists of variable and constant regions divided into CH1, CH2,
and CH3 domains.
● CH2 domain includes the complement-binding site.
● CH3 domain is where IgG attaches to receptors on neutrophils and macrophages.
Antigen-Binding Site Specificity:
● Determined by the amino acid sequence of the hypervariable region.
Understanding antibody structure is crucial for comprehending their diverse roles in the
immune system.


Five Major Types of Antibodies: Antibodies are classified into several types or classes,
each with distinct roles in the immune response. The five major types of antibodies include:
​ IgG (Immunoglobulin G): IgG is the most abundant antibody class in the
bloodstream, accounting for approximately 75-80% of all antibodies. It is highly
versatile and is involved in a broad range of immune functions, including neutralizing
pathogens, opsonization (marking pathogens for phagocytosis), and activating
complement proteins. IgG can cross the placenta, providing passive immunity to the
fetus.
​ IgM (Immunoglobulin M): IgM is the first antibody class produced during an initial
immune response. It is large and primarily found in the bloodstream. IgM is efficient
at agglutinating (clumping) pathogens, making them easier to clear from the body. It
is also a critical component of the B-cell receptor complex.
​ IgA (Immunoglobulin A): IgA is predominantly found in mucosal areas, such as the
respiratory and gastrointestinal tracts. It plays a vital role in mucosal immunity by
preventing pathogen attachment to mucosal surfaces and protecting against
infections. IgA is often found in secretions like saliva, tears, and breast milk.
​ IgE (Immunoglobulin E): IgE is associated with allergic reactions and immune
responses against parasitic infections. It binds to mast cells and basophils, triggering
the release of histamines and other inflammatory mediators when exposed to
allergens. This causes allergic symptoms, such as itching and swelling.
​ IgD (Immunoglobulin D): IgD is primarily found on the surface of B cells as a
receptor. It plays a role in B cell activation and maturation. IgD is less common in
circulation, and its specific functions are not as well understood as the other antibody
classes.

2. Describe the structure of an antibody monomer and state what part of it binds to
Antigens.
● Antibody Monomer Structure:
● Antibodies, also known as immunoglobulins (Ig), have a monomeric structure.
● Each antibody monomer is shaped like a "Y."
● The Y-shaped structure consists of two identical heavy chains and two
identical light chains.
● Binding Site for Antigens:
● The part of the antibody responsible for binding to antigens (foreign
substances) is located at the tips of the "Y."
● This region is referred to as the "antigen-binding site" or "paratope."
● The antigen-binding site is composed of the variable regions of both the
heavy chain and the light chain.
● Within these variable regions, there are three hypervariable regions called
CDRs (complementarity-determining regions).
● The CDRs, especially CDR3 at the junction of the V and C regions, exhibit the
greatest variability in amino acid sequences.
● This diversity allows antibodies to recognize and bind to a wide range of
specific antigens.
● The binding of an antibody to its specific antigen initiates immune responses,
including antigen neutralization, opsonization, and immune cell activation.
Remember that the specific amino acid sequences in the CDRs of the variable regions
determine the antibody's unique ability to recognize and bind to specific antigens.

3. Name and explain the functions of the Fab and Fc fragments of immunoglobulins.
The Fab (Fragment Antigen-Binding) and Fc (Fragment, Crystallizable) fragments of
immunoglobulins have distinct functions:
​ Fab Fragment (Fragment Antigen-Binding):
● Function: Binds to antigens (foreign substances, pathogens).
● Composition: Contains one constant and one variable domain from both
heavy and light chains.
● Specific Role: Shapes the antigen-binding site, also known as the paratope,
located at the amino terminal end of the monomer.
● Mechanism: The two variable domains, one from each heavy and light chain,
interact with specific epitopes on their corresponding antigens.
● Result: This binding initiates immune responses against the recognized
antigens.
​ Fc Fragment (Fragment, Crystallizable):
● Function: Modulates immune cell activity and mediates various physiological
effects.
● Composition: Comprises two heavy chains, contributing two or three constant
domains, depending on the antibody class.
● Key Roles:
● Protein Binding: Binds to specific proteins, ensuring the appropriate
immune response for a given antigen.
● Cell Receptor Interaction: Interacts with various cell receptors, such as
Fc receptors on immune cells.
● Interaction with Immune Molecules: Also binds to other immune
molecules, including complement proteins.
● Physiological Effects: Mediates effects like opsonization (enhancing
phagocytosis), cell lysis (cell destruction), and degranulation of
immune cells like mast cells, basophils, and eosinophils.
5. List and describe features of the structure and functions of the major classes of
Immunoglobulins.
6. Describe ways in which an antibody acts against an antigen.
Neutralization: Antibodies can bind to antigens, such as viruses or toxins, and block their
harmful effects. By neutralizing these antigens, antibodies prevent them from infecting cells
or damaging tissues.
Opsonization: Antibodies can enhance the process of phagocytosis, where immune cells
called phagocytes (e.g., macrophages and neutrophils) engulf and destroy pathogens.
Antibodies attach to antigens, making them more visible to phagocytes and improving the
efficiency of pathogen clearance.
Complement Activation: Antibodies can trigger the complement system, a group of
proteins that form pores in the membranes of pathogens. This process leads to cell lysis
(bursting) and the destruction of the pathogen.
7. Provide examples of using monoclonal antibodies in therapy.

You might also like