Вы находитесь на странице: 1из 31

Ch.

5 The Complement System

VIDEO 1: What is complement?


“The complement system is a set of proteins that circulates in the blood serum. When
activated, these proteins work together to destroy foreign cells by cytolysis, to activate the
inflammatory response and to assist in phagocytosis. The main components of the
complement system are called C1 through C9.”

VIDEO 2: Complement Activation: 3 Pathways


“Complement can be activated through three different pathways: the classical
pathway, the alternative pathway, and the lectin pathway.
The classical pathway was the first activation pathway of the complement system to
be discovered. The classical pathway begins when a pair of antibodies attached to antigens,
such as proteins or a large polysaccharide on the surface of a bacterium. The antibodies bind
and activate C1. C1 then cleaves several copies of C2 and C4. Together, the C2a and C4b
fragments form an enzyme that cleaves C3, activating the complement system.
The alternative pathway of complement activation was so named because it was
discovered after the classical pathway. While the classical pathway requires antibodies
specific to the invading microorganism, the alternative pathway requires 3 complement
proteins called factor B, factor D, and factor P (or properdin). These products are attracted
to microbial cell-surface material such as the lipid-carbohydrate complexes of certain
bacteria and fungi. Together with C3, these proteins result in the cleavage and activation of
C3 proteins, which in turn activate the rest of the complement system.
The most recently discovered pathway of complement activation is the lectin
pathway. When macrophages ingest material through phagocytosis, they release chemicals
that stimulate the production of carbohydrate-binding proteins called lectins. The lectins bind
to distinctive patterns of carbohydrates on the surfaces of certain bacteria and viruses. The
lectins themselves can act as opsonins for phagocytosis, but also activate C2 and C4, leading
to the activation of C3, which leads to the activation of the rest of the complement
pathway.”

VIDEO 3: Complement Functions:


“Once the complement cascade is activated, several events occur that ultimately
result in the destruction of the pathogen. C3a and C5a act as chemotactic factors, attracting
phagocytes to the site of infection. C3a and C5a also bind to mast cells, triggering the release
of histamine and other chemicals as part of inflammation. C3b acts as an opsonin, coating the
invading cell. This coat makes it easier for the phagocyte to adhere to the invading cell during
phagocytosis. C5b binds to C6 and C7, which attach to the plasma membrane of an invading
cell. C8 and several C9 molecules bind to the C5b-C6-C7 complex, forming a cylinder-shaped
membrane attack complex (or MAC). The MACs form circular holes in the membrane of the
invading pathogen, causing water to enter the cell and ions to leave. This results in cytolysis,
killing the cell.”

The complement system


● Group of serum proteins circulating in inactive form
● Once activated, there are multiple possible outcomes:
○ Target cell membrane lysis: rupture of membranes due to formation of
membrane attack complex.
○ Chemotaxis: attract macrophages and neutrophils by inflammatory mediators,
C5a, and to a lesser extent C3a and C4a.
○ Opsonization: enhances phagocytosis by opsonins (C4b and C3b) binding to
foreign antigens.
○ Inflammation
○ Agglutination: causes clustering and binding of pathogens.

Three complement activation pathways exist:


● Classical
● Lectin
● Alternative

All three generate C3b, an important, multifunctional complement protein. With the help of
other complement proteins, the signal is amplified and there are a variety of functional
consequences such as inflammation, opsonization, lysis, or inflammation (see picture on next
page)
Classical Pathway:
● IgG binds to a multivalent antigen (the antigen has several
sites where the antibody can attach)
● This allows binding of C1q, beginning the process of
complement deposition
● C1 finds Fc on adjacent IgG molecules bound to the antigen
● The classical pathway is initiated by this antibody binding
○ C1qrs initiates a cascade of reactions enabling the
next reaction in the sequence
○ C1 binding is followed by cleavage of C4, then C2
○ C4b and C2a bound to the cell surface make up an
enzyme called C3 convertase
○ C3 convertase cleaves many C3 proteins
○ Some C3bs combine with C3 convertase to form C5 convertase
○ C5 convertase cleaves C5 protein
Above: (THIS IS MORE DETAILED than the prior explanation)
1. C1q binds antigen-bound antibody and induces a conformational change in one C1r
molecules, activating it. This C1r then activates the second C1r and two C1s molecules
2. C1s cleaves C4 and C2. C4 is cleaved first and C4b binds to the membrane close to C1.
C4b binds C2 and exposes it to the action of C1s. C1s cleaves C2, creating the C3
convertase (C4b2a)
3. C3 convertase hydrolyses many C3 molecules. Some combine with C3 convertase to
form C5 convertase
4. The C3b component of C5 convertase binds C5, permitting C4b2a to cleave C5

Lectin Pathway
● The lectin pathway is initiated when soluble proteins
recognize microbial antigens
● Lectins (AKA mannose-binding lectin, AKA MBL) bind
to microbial surfaces
● Lectins can serve as docking sites for MBL-associated
serine proteases (MASPs)
○ MASPs cleave C4 and C2 to form the C3
convertase
○ Subsequent steps are the same as the classical
pathway.

Alternative Pathway
● Initiated in 3 ways
○ Activated C3b binds to membrane of target cell
■ Factor B binds and is cleaved by factor D
■ C3bBb at the membrane is the C3 convertase
■ Properdin (AKA factor P) stabilizes the C3 convertase

○The alternative properdin-


activated pathway
■Properdin can directly bind to a
surface
■ This can then recruit C3b and Factor B
■ Factor D is recruited and cleaves Factor B into Bb
■ The resultant C3bBb is an active C3 convertase
■ Subsequent steps are the same as the alternative tickover pathway.

○The alternative protease-


activated pathway
■ Initiation of clotting cascades has also been shown to stimulate cleavage
of complement proteins
● Thrombin cleaves C3 and C5 in vitro
● Platelet activation releases ATP, Ca ions, and serine/threonine
kinases that could stabilize C3b in fluid phase
■ This indicates that strong inflammatory reactions could potentially
activate complement systems.
● The three pathways converge at the formation of C5 convertase
● C5 initiates the generation of the membrane attack complex (MAC)
○ The MAC is the result of deposition of C5b, C6, C7, C8, and C9 in target cell
membranes
○ This pore structure disrupts osmotic integrity, resulting in cell death.

The diverse functions of complement


● Complement receptors connect complement-tagged pathogens to effector cells
○ CR1 on leukocytes and erythrocytes
■ On erythrocytes, it helps to bring immune complexes to the liver for
clearance by phagocytes
■ On phagocytes, it helps to bind complement-coated bacteria to enhance
ingestion and destruction
■ On B cells, it helps bind to complement-coated antigens, enhancing
ingestion for processing and presentation to helper T cells.
○ C3aR/C5aR on granulocytes
■ Stimulates release of proinflammatory cytokines and granule
components from basophils, eosinophils, and neutrophils
● Receptors on host cells allow for discrete and differentiated
responses.
● Complement enhances host defense against infection
○ MAC-induced cell death
○ Promotion of inflammation
○ Promotion of opsonization
■ Opsonized microbes are easier to ingest/
destroy (below, left)
■ Opsonized immune complexes are easier to
clear (right)

● Complement mediates
interface between innate and adaptive immunity
○Enhances antigen uptake of antigen
bound to MBL, C1q, C3b, and C4b that bind to
receptors on APCs
○ Enhances B-cell response by increasing avidity of B-cell binding to complement-
bound antigen
○ Lyse immature T cells with low sialic acid content (a carbohydrate that
increases in concentration as a protective coating on maturing T cells)
○ Binding of C3a, C5a, and C3b to their receptors on mature T cells facilitates
their growth, differentiation, and survival.
● Complement aids in the contraction phase of the immune response
○ As lymphocytes are no longer required, complement aids in disposal of
apoptotic cells and bodies.
○ Complement also aids in removal/disposal of immune complexes formed during
responses
○ These responses avoid damaging inflammation induction in the absence of
antigens following clearance of an infection.
● Complement activity is passively regulated by protein stability and cell surface
composition
○ Short half-life of C3 convertase unless stabilized by properdin
○ Self-cells possess different carbohydrate structures that are more effectively
bound by fluid phase proteases
■ These more readily inactivate C3b through hydrolysis, protecting self
cells

The regulation of complement activity


● Numerous regulatory proteins help to prevent the complement system from harming
self-cells
● The C1 inhibitor, C1INH, promotes dissociation of C1 components
○ Binds in the active site of serine proteases
○ Causes C1r2s2 to dissociate from C1q

○ No further cleavage of C4 or C2 is possible


○ Inhibits initiation of classical and lectin complement pathways.
● Decay accelerating factors promote decay of C3 convertases
○ Several different proteins with similar activities
■ DAF (CD55), CR1, C4BP (C4 binding protein)
■ Factor H binds negatively charged cell surface sialic acid and heparin,
molecules unique to eukaryotic cell surfaces
● Work to accelerate decay of C4b2a (C3 convertase) on the surface of host cells. (see
below)
● Factor I degrades C3b and C4b
○ Soluble, constitutively active serine protease
○ Cleaves membrane-associated C3b and C4b into inactive fragments
■ Requires MCP (CD46) and CR1 (found on membranes of host cells) to
function

● Protectin (CD59) inhibits the MAC attack


○ Binds C5b678 complexes deposited on host cells
■ Prevents their insertion into the plasma membrane
■ Also blocks C9 recruitment, preventing MAC formation
○ Similarly, soluble complement S protein (vitronectin) binds fluid-phase C5b67 to
prevent insertion into host cell plasma membranes

● Carboxypeptidases can inactivate the anaphylatoxins C3a and C5a


○ Remove arginine residues from the C termini of C3a and C5a
■ Creates des-Arg (without arginine) inactive forms
■ Helps to shut down unnecessary or dangerous chemotactic and
inflammation induction.

Complement deficiencies
●Genetic deficiencies have been
described for each of the complement components, but outcomes vary
○ Patients with any C1q, C1r, C1s, C4, or C2 deficiency often present with
immune complex disorders due to inadequate clearance
○ Some with MBL deficiency may exhibit greater frequency of infections by
encapsulated bacteria due to inefficient opsonization and phagocytosis
● Animal models exist for most complement deficiencies, allowing for further study.

Microbial complement evasion strategies


● Different mechanisms exist and are highly varied
○ Some interfere with the first step of Ig-mediated complement activation
○ Microbial proteins may bind and inactivate complement proteins
○ Microbial proteases destroy complement proteins
○ Some microbes mimic or bind complement regulatory proteins.

The evolutionary origins of the complement system


● Five families of genes for complement components
○ Complement served to assist phagocytosis prior to the evolution of adaptive
immunity

Summary
● The complement system serves many different purposes, helping to link innate and
adaptive immune responses
● It is tightly controlled but enhances many other responses once activated
● It also provides a window into the evolution of immunity
● Understanding the system and its methods of activation and regulation helps us to
better understand innate immunity and evolution.

Ch. 4 (cont.)

Induced cellular innate responses


● PRR signaling pathways activate expression of a large variety of genes
○ Type I interferons (potent antiviral effects)
○ 4 genes are turned on by IFN
■ Protein kinase R (PKR: RNA activated)
■ 2’, 5’-oligoadenylate synthetase (an antiviral enzyme that counteracts
viral attack by degrading viral and host RNA)
■ Mx group proteins (inhibit negative-stranded RNA viruses) serve as PRRs
■ IFIT (interferon induced proteins with tetratricopeptide repeats)
■ Explanation below:
Phagocytosis
● Defined as engulfment and internalization of materials such as microbes for their
clearance and destruction
● Microbes are recognized by receptors on phagocytes
● They may also recognize PAMPs directly

Human receptors that trigger phagocytosis:


Clinical Correlation: Human Cancers
● Cancer cells with high CD47 expression are anti-phagocytic (evade phagocytosis)
○ Blockade of CD47 production increases phagocytosis of cancerous cells and
leaves most normal cells unaffected
● Cancers with high calreticulin expression are pro-phagocytic;
○ blockade of calreticulin/LRP interaction prevents anti-CD47 antibody mediated
phagocytosis.
If you’re still confused read this: https://www.ncbi.nlm.nih.gov/pubmed/21178137

Microbes are recognized by receptors on phagocytes:


● May recognize soluble opsonin protein bound to microbes
● Opsonin= a molecule that promotes phagocytosis by binding to a microbe (Eg.
mannose binding lectin, or MBL)

Complement pathways: Highly recommend just reviewing Ch. 5 b/c Dr. J’s explanation of
complement is not super great.
Ingested materials are taken into phagosomes
● Phagosomes are fused with lysosomes or granules
○ Destruction occurs through enzyme degradation, antimicrobial proteins, and
toxic effects of reactive oxygen and reactive nitrogen species (ROS and RNS)
○ Known as “respiratory burst”

Regulated cell death (Apoptosis)


● Cell death is induced by receptor-activated signal pathways
● Apoptosis is induced by TNF binding to TNFR (present on NK cells and cytotoxic T cells)
● Neutrophil extracellular trap (NETs) activation requires NADPH oxidase and generation
of ROS
● Pyroptosis, induced by inflammasome activation, eliminates infected macrophages
allowing release of IL-1β and IL-18

Inflammatory responses:
● Early components of inflammation include:
○ Increased vascular permeability
○ Recruitment of neutrophils and other leukocytes from the blood to the site of
damage/infection
○ Later stages of inflammation are due to the acute phase responses (APRs)
■ Induced by proinflammatory cytokines (IL-1, TNF-α, IL-6)

Tissue damage by bacteria:


Exudate
Extravasation
Attraction of
phagocytes
NK cells
NK cells are lymphocytes with innate immune functions
● Express a limited set of receptors: Killer activation receptors (KAR) and Killer
inhibitory receptors (KIR)
○ Infections
○ Malignant transformations
○ Other stresses
● Activated NK cells perform one of two functions after binding to KARs
○ Kill the altered self-cell by releasing perforin and granzymes inducing apoptosis
○ Produce cytokines that induce adaptive responses against the altered self-cell

Evasion of innate and inflammatory responses


Pathogens have evolved strategies to block, evade, and escape these responses
● Defects in PRRs and signaling pathways increase susceptibility to infections
● Defects that allow the systems to remain abnormally “turned on” contribute to
inflammatory disorders.

Interactions between innate and adaptive immune systems:


A constant interplay between the two systems exists.
● Several innate systems have been co-opted by adaptive immunity to contribute to
antibody-mediated pathogen elimination
○ Opsonization
○ Complement activation
● Some lymphocytes express TLRs, but use them as costimulatory receptors
● Dendritic cells are a key bridge.
○ They bring antigens from the site of infection and present them to T cells in
the lymph nodes
○ This activates the T cells, allowing them to differentiate into particular
pathogen-specific subsets for the best antigen clearance (TH cell subsets and
TC cells)

Above:
A) Signal one determines antigen specificity and consists of interaction of the TCR with
peptides loaded onto DC MHC molecules. Signal two consists of co-signaling and can be
either positive, leading to cell activation (co-stimulation) or negative, leading to no
response (co-inhibition). Co-signaling molecules, CD80 and CD86, are upregulated on
DC after binding of PAMPs to their cognate receptors.
B) Signal three involves the polarization of CD4 T cells into either Th1, Th2, or regulatory
T cells. Immature DC are polarized by the binding of PAMP and differentiate into
mature DC that induce the formation of Th1, Th2, or Treg cells, respectively.
a) In general, viral associated PAMPs give rise to Th1 responses, and PAMPs from
parasitic organisms favor Th2 responses.
C) Signal four leads to spatial imprinting of T cells, leading to the acquisition of homing
receptors that induce selective recirculation through the tissue in which antigen was
first encountered.
a) Eg. DC from the intestine uniquely produce retinoic acid, inducing T cells to
upregulate the T cell gut-homing receptors a4b7 and CCR9 and suppress
expression of the skin-homing receptor cutaneous lymphocytes antigen (CLA).

Ubiquity of innate immunity


Innate immunity is evolutionarily “older” than adaptive immunity
● TLRs are unique to animals
● PRRs with leucine-rich repeats (LRRs) are found in virtually ALL plants and animals

Summary
● Basic barrer mechanisms and simple biochemical defenses are the frontline of
immunity
● Innate immune responses depend on recognition of general pathogen molecules
○ Responses are varied, but include:
■ Phagocytosis
■ Triggering of inflammatory responses
■ Direct destruction by NK cells
■ Initiation of adaptive immune responses
● Learning the first defense strategies of immunity helps to better understand later
adaptive methods.

Ch. 7 MHC and Antigen Presentation

VIDEO 1: A critical function of antigen presenting cells:


“Certain phagocytic cells of the immune system, called antigen-presenting cells, seek out
foreign cells and engulf them. In addition, despite the best efforts of the immune system,
cells can become infected. In both situations, the cell needs a way to communicate with
immune system cells. The phagocyte needs to show what it has captured. The virus-infected
cell needs to show that it is infected. Antigen processing and presentation is a way for a cell
to display information about its activities.”

VIDEO 2: Specialized antigen presenting cells present antigens complexed with class I and II
MHC
“APCs such as macrophages and DCs have two classes of special proteins that are used to
present antigens on their surfaces. Collectively, these proteins are called MHC, which stands
for major histocompatibility complex. Class I MHC proteins are used to present antigens
produced within the cell. A difference class of MHC proteins, class II MHC proteins, are used
to present antigens engulfed by APCs. Most of the other cells in the body, including infected
cells, are also able to present antigens. However, these cells only have the Class I MHC
proteins.”
VIDEO 3: Special APCs present pathogenic/endogenous peptides to T cells.
“Special APCs including monocytes, macrophages and their relatives engulf invading
pathogens and digest them, producing fragments with antigenic determinants. Vesicles
containing MHC proteins fuse with vesicles containing digested pathogens. The antigenic
determinants from the pathogens are loaded onto the MHC proteins. MHC proteins are used to
present peptides on the surface of the cell. Since the peptides are produced from proteins
inside the cell, they allow cells of the immune system to detect proteins inside the cell
without entering the cell. Cells in the body routinely break down proteins into small peptides
which are about 8-10 AA in length. These peptides enter the ER, and are loaded onto MHC
proteins. The vesicles containing the MHC-peptide complexes bud off the ER, and are released
into the cytosol and travel to the cytoplasmic membrane, delivering the complexes to the
exterior of the cell. If this cell is functioning normally, the immune system cells will simply
ignore this presentation. However, if the cell is infected and thus presenting viral proteins,
the immune system cells can notice the problem and take action”

VIDEO 4: Helper and Cytotoxic T cells destroy infected/abnormal cells


“Cell-mediated immunity is a host defense used to fight intracellular pathogens and abnormal
body cells, such as cancer cells. The protagonists of cell mediated immunity are T-
lymphocytes, which can be divided into two groups: helter T cells and cytotoxic T cells.
Helper T cells secrete cytokines to activate other cells of the immune system, while cytotoxic
T cells attack infected cells directly.”

Structure and function of MHC molecules


Class I molecules
● 45 kDa glycoprotein α chain
● 12 kDa β2-microglobulin protein
● α chain passes through plasma membrane
● α1 and α2 domain α-helix and β-pleated sheet form walls and floor of peptide binding
site, respectively.
Class II molecules
● Member of the Ig superfamily
● Possesses Ig domains
● Heterodimeric
○ A 33kDa α chain
○ A 28 kDa β chain
● Both chains pass through the plasma membrane
● A peptide-binding cleft is formed by the pairing of the α1 and β1 domains
○ Accommodates peptides of 13-18 AA in length.

Class I and II molecules exhibit polymorphism in the peptide-binding region


● Several hundred different allelic variants in humans
○ Up to 6 class I and about 12 class II molecules expressed per person
○ How does this limited group of molecules present the vast diverse array of
possible antigen peptide fragments?
■ A given MHC molecule can bind numerous different peptides, and some
peptides can bind to several different MHC molecules.
● Class I MHC-peptide interactions
○ Present peptides to CD8+ T cells
○ Peptides derived from endogenous intracellular proteins
○ Some AA anchor the peptide into the groove
○ Other AA are available to interact with a TCR.

General organization and inheritance of MHC


● Class I and II molecules exhibit diversity at individual and species levels
● Individuals express MHC alleles inherited from both parents
● Alleles can also differ among individuals, creating enormous diversity
● This diversity provides flexibility in responding to unexpected environmental changes,
now and in the future

Role of MHC and expression patterns


● MHC molecules present both intracellular and extracellular antigens
● Class I presents intracellular antigen peptides
○ Includes self-proteins
○ Provides a way for checking that cells are self and are generally healthy
○ Can show which cells have been infected with viruses or are abnormal and
display those peptides on class I and activate Tc cells.
● Class II presents extracellular antigen peptides
○ More restricted, generally found on cells involved in immune responses
○ Helps to direct responses against threats-- things that shouldn’t be in our
systems-- and display those peptides on class II and activate Th cells.
● MHC class I expression is found throughout the body
○ Essentially all nucleated cells
● MHC class II is primarily restricted to antigen-presenting cells, macrophages, B cells,
and dendritic cells.

Antigen-presenting cell types

● MHC expression can change with changing conditions


○ Genetic regulatory components
■ Promoters that drive up transcription during times of infection, NLRs are
core component transcriptional activators
○ Viral interference
■ Viruses like to shut down MHC Class I expression because it targets the
cells they’re in for destruction
○ Cytokine-mediated signaling
■ Some cytokines like IFN-α and TNF-α expressed first during infection/
disease can drive up MHC expression, whereas corticosteroids and
prostaglandins downregulate MHC expression
● MHC alleles play a critical role in immune responsiveness
○ Class II MHC alleles and antigen presentation
■ Different capability to present antigens may dictate overall strength of
immune response from individual to individual.
● T cells are restricted to recognizing peptides presented in the context of self-MHC
alleles
○ CD8/CD4+ T cells can only recognize peptides presented by MHC class I/II,
respectively
○ Whole antigens cannot be recognized by T cells
○ Only processed antigen peptide fragments can be recognized.

Role of MHC and expression patterns:


● Evidence suggests different antigen processing and presentation pathways
○ Class I presentation requires cytosolic or endogenous processing
○ Class II presentation requires exogenous processing. (see below)
Dendritic cell presentation of antigen to CD4+ helper T cell

Dendritic cell cross-presentation of antigens to CD4+ helper T and cytotoxic T cells

SUMMARY: Processing and presentation of antigens:


1.Ingestion of antigen by the APC by phagocytosis
2. Digestion of antigen into fragments
3. Synthesis of MHC molecules
4. Fusion of vesicles containing antigen fragments and MHC proteins
5. Binding of fragments to MHC molecules
6. Insertion of antigen-MHC complex into the plasma membrane

Why is antigen processing/presentation necessary for adaptive immune response?


● T cells need to have antigen peptides presented to them for activation
● Understanding the molecules that perform this presentation is crucial to understanding
T cell activation
● Also, understanding how intracellular and extracellular antigens are processed for
presentation gives us insight into why different T cells are stimulated/activated during
immune responses

CH. ? Adaptive Immunity (I’m pretty sure we had an identical lecture in microbio)

Overview: Adaptive immunity is the body’s ability to recognize and defend itself against
distinct invaders and their products

Five attributes of adaptive immunity:


● Specificity
● Inducibility
● Clonality
● Unresponsiveness to self
● Memory
Two types of lymphocytes
● B lymphocytes (B cells) mature in the bone marrow
● T lymphocytes (T cells) mature in the thymus
Two types of adaptive immune responses
● Humoral immune responses (respond to antigens in the blood)
● Cell mediated immune responses

Elements of Adaptive immunity


● Tissues and organs of the lymphatic system
○ Lymphoid organs
■ Primary lymphoid organs: red bone marrow, thymus
■ Secondary lymphoid organs: Lymph nodes, spleen, tonsils, MALT.
○ These screen the tissues of the body for foreign antigens
○ Composed of lymphatic vessels and lymphatic cells, tissues, and organs
○ One way system that conducts lymph from tissues and returns it to the
circulatory system.
● Antigens
○ Molecules the body recognizes as foreign and worthy of attack
○ Recognized by 3D regions called epitopes (AKA antigenic determinants). (See
picture).
○ Include various bacterial components as well as proteins of viruses, fungi, and
protozoa
○ Food and dust can also contain antigenic particles.

Humoral Immune responses


● Humoral immune responses are mounted against exogenous pathogens
● Activates only in response to specific pathogens
● Can generate antibodies to millions of antigens (VDJ recombination and
hypermutation)

Back to elements of adaptive immunity:


● B lymphocytes and antibodies
○ Arise and mature in the red bone marrow
○ Found primarily in the spleen, lymph nodes and MALT
○ Small percentage of B cells circulate in the blood
○ Major function is secretion of antibodies.
○ Specificity of the BCR (B cell receptor)
■ Each B lymphocyte has multiple copies of the B cell receptor
■ Each B cell generates a single BCR
■ Two variable regions of the BCR form the antigen-binding sites
■ Each BCR recognizes only one epitope
■ The entire repertoire of an individual’s BCR is capable of recognizing
millions of different epitopes.

Activation leads to secretion of membrane bound


antibodies

○Specificity and antibody structure


■Ab are immunoglobulins similar to BCRs
■ Secreted by activated B cells called plasma cells
■ Have identical antigen-binding sites and antigen specificity as the BCR
of the activated B cell

Binding of a T-independent antigen by a B cell:


T-dependent humoral immune response:

Inducement of T-dependent humoral immunity


● Plasma cells
○ Majority of cells produced during B cell proliferation
○ Only secrete antibody molecules complementary to the specific antigen
○ Short lived cells that die within a few days of activation
■ Their antibodies and progeny can persist.

Isotype switching or class switching is a biological mechanism that changes a B cell’s


production of immunoglobulin from one type to another, such as from the isotype IgM to the
isotype IgG. During this process, the constant region portion of the antibody heavy chain is
changed, but the variable region of the heavy chain stays the same. Since the variable
region (Fab region) does not change, class switching does not affect antigen specificity.

Antibody function
● Antigen-binding sites are complementary to epitopes
● Antibodies function in several ways
○ Activation of complement and inflammation
○ Neutralization
○ Opsonization
○ Killing by oxidation
○ Agglutination
○ Antibody-dependent cellular cytotoxicity (ADCC)

Classes of antibodies
●Threats confronting the
immune system are
variable
● Class involved in the immune response depends on the type of antigen, portal of entry,
and antibody function needed
● 5 different classes:
○ IgM: the first Ab produced
○ IgG: most common and longest-lasting
○ IgA: associated with body secretions
○ IgE: involved in response to parasitic infections and allergies.
○ IgD: exact function unknown

Memory B cells and establishment of immunological memory


● Produced by B cell proliferation but do not secrete antibodies
● Have BCRs complementary to the antigenic determinant that triggered their
production
● Long lived cells persist in the lymphoid tissue
● Initiates antibody production if antigen is encountered again

Back to elements of adaptive immunity:


● T cells
○ Produced in red bone marrow and mature in thymus
○ Circulate in the lymph and blood and migrate to lymph nodes, spleen, and
Peyer’s patches
○ Antigen-binding sites are complementary to epitopes.
○ Have TCRs on their cytoplasmic membranes
○ Specificity
■ TCRs do NOT recognize epitopes directly
■ TCRs only bind epitopes associated with an MHC protein
■ TCRs act primarily against cells that harbor intracellular pathogens.

● Types of T lymphocytes
○ Based on surface glycoproteins and characteristic functions
○ 3 types
○ Cytotoxic T lymphocytes (CD3 and CD8+) directly kill other cells
○ Helper T lymphocytes (CD3 and CD4+) help regulate activities of B cells and
cytotoxic T cells
○ Regulatory T lymphocytes (CD3, CD4, CD25R, FoxP3+) repress adaptive
immune responses (particularly T cells).
● T cell activation and the two signal hypothesis
○ T cells require antigen presentation as a first signal (MHC + antigen)
○ Other molecular interactions (costimulatory molecules) can provide the second
required activation signal
○ Once activated, T cells differentiate into their effector forms
■ CD8+ T cells go on to become killer T cells
■ CD4+ T cells differentiate into several different subsets
● Focus primarily on activation overall, followed by differentiation
of CD4+ cells.

● Costimulatory signals are required for optimal T cell activation and proliferation
○ Signal 1: antigen specific TCR engagement
○ Signal 2: contact with costimulatory ligands
○ Signal 3: cytokines direct T cell differentiation into distinct effector cell
types (AKA polarization of the T cells into Th1 or Th2)
○ Signal 4: where it will go to respond
Clonal deletion
● Vital that immune responses are not directed against autoantigens
● The body eliminates self-reactive lymphocytes
○ Lymphocytes that react to autoantigens → apoptosis

Back to elements of adaptive immunity:


● Immune response cytokines
○ Soluble regulatory proteins that act as intercellular signals
○ Cytokines secreted by various leukocytes
○ Cytokine network
■ The complex web of signals among all the cells of the immune system.
○ Interleukins (ILs) signal among leukocytes
○ Interferons (IFNs) are antiviral proteins that may act as cytokines
○ Growth factors stimulate stem cells to divide
○ Tumor necrosis factor (TNF) is secreted by macrophages and T cells to kill
tumor cells and regulate immune responses and inflammation
○ Chemokines: chemotactic cytokines that signal leukocytes to move

Cell-mediated immune responses


● Respond to intracellular pathogens and abnormal body cells
● Most common intracellular pathogens are viruses
● The response is also effective against cancer cells, intracellular protozoa and
intracellular bacteria.
● Activation of T cell clones and their functions:
○ Antigen presentation
○ Helper T cell differentiation
○ Clonal expansion
○ Self-stimulation
● Memory T cells
○ Some activated T cells become memory T cells
○ Persist for months or years in lymphoid tissues
○ Immediately functional upon subsequent contacts with epitope specific to its
TCR
● T cell regulation
○ Regulation is needed to prevent T cell response to autoantigens
○ T cells require additional signals from an antigen presenting cell
■ Interaction of the T cell and APC stimulates the T cell to respond to the
antigen.
● What are the differences between natural Tregs and inducible Tregs?
○ Natural Tregs (nTregs) originate from the thymus as CD4 and CD25+ cells
together with expression of the transcription factor FoxP3 and are not really
influenced by the cytokine milieu
■ Natural T regs represent approximately 5-10% of the total CD4+ T cell
population
○ Inducible Tregs (iTregs) originate as CD4+ single positive cells from the thymus
■ Following adequate antigenic stimulation in the presence of cognate
antigen and specialized immunoregulatory cytokines such as TGF-b,
IL-10, and IL-4, differentiate into CD25+ and FoxP3+ expressing
Tregs, hence, they are called “adaptive” or “inducible” since they are
modified by cytokines.

Types of Acquired immunity


● Specific immunity acquired during and individual’s life
● Two types
○ Naturally acquired
■ Response against antigens encountered in daily life
○ Artificially acquired
■ Responses to antigens via vaccine introduction (generate memory cells)
● Either active or passive
○ Passive is when you receive antibodies from another individual.

Вам также может понравиться