flashcards 2

(602 cards)

1
Q

What are the two main lineages of immune cells?

A

They are the myeloid and lymphoid lineages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Where do immune cells originate?

A

They develop from self-renewing stem cells in the bone marrow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

How do stem cells in the bone marrow differentiate?

A

They first become pluripotent stem cells and then progenitor cells, which give rise to various immune cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Which cells are produced from the myeloid lineage?

A

The myeloid lineage produces granulocytes (neutrophils, eosinophils, basophils), mast cells, monocytes/macrophages, dendritic cells, and megakaryocytes (which form platelets).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the role of neutrophils in the immune system?

A

Neutrophils are first responders that are highly phagocytic, secrete NETs, and have a short lifespan (2–3 days).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

How can neutrophils be identified morphologically?

A

They have neutral-staining (pink) cytoplasmic granules and a lobulated nucleus.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What function do eosinophils serve?

A

Eosinophils combat parasitic (helminth) infections and release inflammatory mediators.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is a key characteristic of eosinophils’ granules?

A

Their granules stain bright red and contain major basic protein (MBP).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is the primary role of basophils?

A

Basophils release substances that promote inflammation during allergic responses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How common are basophils in the bloodstream?

A

They account for less than 0.2% of white blood cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What do mast cells do once they mature in tissues?

A

They release histamine and other mediators during allergic reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Which receptor is important on mast cells?

A

They have receptors for IgE antibodies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

How do monocytes differ from macrophages?

A

Monocytes circulate in blood for 1–2 days, then migrate into tissues where they differentiate into larger, adherent macrophages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the main functions of macrophages?

A

They perform phagocytosis, present antigens, and can live for months or years in tissues.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What role do dendritic cells play?

A

They capture antigens in tissues and present them to T cells in lymphoid organs to activate adaptive responses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How are platelets produced?

A

Platelets are produced by the fragmentation of polyploid megakaryocytes in the bone marrow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the main function of platelets?

A

They play a crucial role in blood clotting and hemostasis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What distinguishes lymphocytes in peripheral blood?

A

Lymphocytes are small cells (6–10 microns) with a large nucleus and small cytoplasmic halo, forming 20–30% of white blood cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are the main roles of B lymphocytes?

A

B cells mature in the bone marrow, express markers like CD19/CD20, and differentiate into plasma cells (for antibody production) or memory cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What do plasma cells do?

A

They produce specific antibodies to target pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Where do T lymphocytes mature?

A

T cells mature in the thymus after originating from the bone marrow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What surface markers do T cells express?

A

T cells express the T cell receptor (TCR) along with CD3; helper T cells express CD4, and cytotoxic T cells express CD8.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the function of helper T cells?

A

They secrete cytokines that enhance the functions of B cells, cytotoxic T cells, and macrophages.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

How do cytotoxic T cells eliminate infected cells?

A

They release perforins and granzymes to kill virus-infected or tumor cells and form memory cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
What is the role of regulatory T cells (Tregs)?
Tregs suppress excessive immune responses and help maintain tolerance to self-antigens to prevent autoimmunity.
26
What are γδ (gamma delta) T cells?
They are T cells with TCRs composed of gamma and delta chains, found mainly in mucosal tissues, and recognize lipid antigens.
27
What function do natural killer (NK) cells serve?
NK cells kill virus-infected or tumor cells using receptors other than the T cell receptor.
28
What are innate lymphoid cells (ILCs)?
ILCs help bridge innate and adaptive immunity by providing rapid defense at mucosal surfaces.
29
What distinguishes natural killer T (NKT) cells from conventional T cells?
NKT cells express a T cell receptor (TCR) and CD3 but lack CD4 and CD8, exhibiting properties of both NK and T cells.
30
How are immune cells typically identified in the laboratory?
By their size, granularity, and the expression of specific surface markers (e.g., CD markers), often via flow cytometry.
31
How do growth factors and cytokines influence immune cell development?
They direct the differentiation of pluripotent stem cells into specific immune cell types by signaling through various pathways.
32
Why is antigen presentation important in the immune response?
It activates T cells by displaying processed antigens on MHC molecules, thereby linking innate and adaptive immunity.
33
How do innate and adaptive immune cells work together?
Innate cells provide immediate defense and antigen presentation, while adaptive cells offer specificity, memory, and regulation to effectively eliminate pathogens.
34
What historical event led to the creation of the CD system for immune cell identification?
In 1982, scientists met in Paris to standardize the naming of immune cells by establishing the Cluster of Differentiation (CD) system, which is still in use today.
35
What percentage of white blood cells are granulocytes, and what proportion of granulocytes are neutrophils?
Granulocytes account for 60–70% of white blood cells, and about 90% of these granulocytes are neutrophils.
36
How do macrophages vary by tissue location?
Macrophages adopt different names and characteristics depending on their tissue location—for example, Kupffer cells in the liver, alveolar macrophages in the lung, microglial cells in the brain, and histiocytes in connective tissue.
37
How do T cell receptors (TCR) and B cell receptors (BCR) differ in antigen recognition?
TCRs recognize short peptide antigens presented by MHC molecules, whereas BCRs bind directly to native antigens (usually larger proteins) and are part of the antibody production process.
38
What role do cytokines and growth factors play in immune cell development?
Cytokines, interleukins, and colony stimulating factors guide the differentiation of pluripotent stem cells into specific immune cell types by signaling through various developmental pathways in the bone marrow and tissues.
39
Which CD markers are commonly used to identify monocytes and macrophages?
Monocytes and macrophages are typically identified using markers such as CD14 and CD15.
40
How are B cell subpopulations characterized?
B cells are divided into subgroups like B1 and B2, and they express surface markers such as CD19 and CD20; upon activation, they differentiate into plasma cells for antibody production or become memory cells.
41
What is the significance of memory cells in the adaptive immune response?
Memory cells, derived from both T and B cells, enable a faster and more effective response when the same pathogen is encountered again.
42
What are some subtypes of helper T cells and their roles?
Helper T cells can be subdivided into TH1 (promote cell-mediated immunity), TH2 (support humoral immunity), TH17 (involved in inflammatory responses), and TReg (regulatory) cells, each secreting distinct cytokines to modulate the immune response.
43
What are the three types of lymphoid tissues?
Primary, secondary, and tertiary lymphoid tissues.
44
What is the function of primary lymphoid tissues?
They are responsible for the development, differentiation, and 'education' of lymphocytes.
45
What are the main primary lymphoid organs?
The thymus (for T cell development) and bone marrow (for B cell development).
46
What is the function of secondary lymphoid tissues?
They allow the accumulation of antigen, its presentation, and interactions with naïve and memory lymphocytes to initiate immune responses.
47
What are the key secondary lymphoid organs?
Lymph nodes, spleen, and mucosa-associated lymphoid tissue (MALT).
48
What are tertiary lymphoid tissues?
They are scattered aggregations of lymphoid cells in tissues, often formed during chronic inflammation, and contain memory lymphocytes.
49
What is the function of the thymus?
It provides an environment for T cell maturation and education.
50
How does the thymus change with age?
It gradually shrinks (involution), becoming less functional in adulthood.
51
What are the two main regions of the thymus?
The outer cortex and the inner medulla.
52
What is the sequence of T cell development in the thymus?
Thymocytes enter as double-negative (no CD4/CD8), become double-positive (expressing both), and then are selected to become either CD4+ helper or CD8+ cytotoxic T cells.
53
How does positive selection occur in the thymus?
Thymocytes that can bind to MHC molecules survive, ensuring they can interact with antigen-presenting cells.
54
How does negative selection work in the thymus?
Thymocytes that bind too strongly to self-peptides are eliminated by apoptosis to prevent autoimmunity.
55
Approximately what percentage of thymocytes survive selection?
Only about 2% of thymocytes successfully mature into naïve T cells.
56
How long does T cell maturation in the thymus take?
Approximately 1–3 weeks.
57
Where do B cells mature?
In the bone marrow.
58
What happens during B cell negative selection?
B cells that bind self-molecules too strongly undergo apoptosis to avoid autoimmunity.
59
What is positive selection in B cell development?
B cells must receive survival signals in the bone marrow to mature properly.
60
What is the lymphatic system?
It is a network of vessels that transports lymph—a fluid containing lymphocytes—between tissues, lymphoid organs, and the bloodstream.
61
What role do lymphatic vessels play?
They drain extracellular fluid, transport immune cells, and facilitate antigen presentation in lymphoid tissues.
62
How do lymphocytes enter tissues from blood vessels?
By binding to adhesion molecules, such as LFA-1 on lymphocytes interacting with ICAM-1 on endothelial cells.
63
What is lymphocyte homing?
It is the directed migration of lymphocytes to specific tissues, guided by adhesion molecules and chemokines.
64
What are lymph nodes?
They are small, bean-shaped organs located at lymphatic junctions that filter antigens and facilitate immune activation.
65
What are the three main regions of a lymph node?
The cortex (B cell follicles), the paracortex (T cells and antigen-presenting cells), and the medulla (plasma cells secreting antibodies).
66
What distinguishes primary follicles from secondary follicles in lymph nodes?
Primary follicles are dense clusters of naïve B cells, while secondary follicles contain germinal centers where B cells proliferate after antigen exposure.
67
How do antigens enter and exit lymph nodes?
They enter via afferent lymphatics and exit via efferent lymphatics that drain into the bloodstream.
68
What is the role of High Endothelial Venules (HEV) in lymph nodes?
HEVs are specialized blood vessels that enable lymphocytes to enter lymph nodes from the bloodstream.
69
What is the function of the spleen?
It filters damaged red blood cells and supports immune responses by housing lymphocytes.
70
What are the two main compartments of the spleen?
The red pulp (which filters aged or damaged red cells) and the white pulp (which contains lymphocytes for immune responses).
71
What is the Periarteriolar Lymphatic Sheath (PALS)?
A T cell–rich region in the spleen that surrounds arterial branches.
72
What is MALT?
Mucosa-associated lymphoid tissue is diffuse lymphoid tissue found in the mucosal linings of the gut, respiratory, and urogenital tracts.
73
What are the two best-characterized types of MALT?
Gut-associated lymphoid tissue (GALT) and bronchus-associated lymphoid tissue (BALT).
74
What is a key feature of Peyer’s patches in the gut?
They contain B cell follicles, T cells, and specialized M cells that transport antigens from the gut lumen.
75
How do antigens enter Peyer’s patches?
Through M cells, which selectively transport antigens from the gut lumen to immune cells.
76
What is unique about MALT compared to other lymphoid tissues?
MALT lacks afferent lymphatics and directly samples antigens from mucosal surfaces.
77
What maintains lymph flow in the lymphatic system?
Arterial pressure, smooth muscle contractions in lymphatic vessels, and backflow prevention valves maintain directional lymph flow.
78
How do leukocytes migrate to sites of infection or inflammation?
They express homing receptors that guide their movement toward chemokine gradients.
79
How do leukocytes migrate to sites of infection or inflammation?
They express homing receptors that bind to adhesion molecules on endothelial cells, directing them to specific tissues.
80
What is gut homing in the context of lymphocyte migration?
It is the process by which lymphocytes target the intestine by expressing receptors that bind to gut-specific endothelial molecules.
81
How do lymphocytes recirculate throughout the body?
They continuously migrate between the blood, lymph nodes, and other secondary lymphoid tissues via the lymphatic system.
82
What are tertiary lymphoid tissues and when do they form?
They are loosely organized aggregates of lymphoid cells that form at sites of chronic inflammation and may contain memory lymphocytes.
83
What is the structure of a thymic lobule?
Each lobule of the thymus has an outer cortex and an inner medulla where thymocyte maturation occurs.
84
What role do thymic nurse cells play?
They assist in the development and maturation of thymocytes within the thymic cortex.
85
What happens to a thymocyte that fails positive selection?
It undergoes apoptosis and is eliminated.
86
What happens to a thymocyte that binds too strongly to self-antigens?
It undergoes negative selection (apoptosis) to prevent potential autoimmunity.
87
How do B cells become functionally mature in the bone marrow?
They undergo both positive selection, responding to survival signals, and negative selection to avoid self-reactivity.
88
How does antigen presentation occur in secondary lymphoid organs?
Dendritic cells and macrophages process and present antigens to T cells, triggering adaptive immune responses.
89
What is innate immunity?
It is the non-specific arm of the immune system that acts as the first line of defense, ready from birth.
90
What triggers the innate immune response?
Tissue damage from trauma or infection triggers the response.
91
What is the primary aim of the innate immune response?
To limit the spread of damage, eliminate microorganisms, and repair tissue damage.
92
Over what time period does the innate immune response typically operate?
It is active from the moment of infection (zero hours) up to about 96 hours.
93
When is the adaptive immune response recruited?
If the pathogen is not controlled within 96 hours, adaptive immunity is recruited.
94
Which cell types are key players in innate immunity?
Phagocytes (neutrophils, monocytes/macrophages, dendritic cells), natural killer (NK) cells, innate lymphoid cells, NKT cells, gamma delta T cells, eosinophils, and B1 cells.
95
What are the four main features (or barriers) of innate immunity?
Anatomical, physiological/chemical, phagocytic/endocytic, and inflammatory barriers.
96
What constitutes the anatomical barrier in innate immunity?
Physical structures like the skin and mucosal surfaces that prevent pathogen entry.
97
How does the skin function as an anatomical barrier?
Its multiple layers (stratum corneum, granular, spinous, basal) and constant shedding of dead cells block microbial entry.
98
What role do commensal microflora play on the skin?
They secrete bacteriocins and metabolites (e.g., lactic acid) that inhibit pathogen growth.
99
What features protect mucosal surfaces?
Mucus, cilia, and secretions (urine, saliva, tears, milk) help clear and prevent microbial invasion.
100
What are the physiological/chemical barriers in innate immunity?
Factors such as normal body temperature, low pH, and antimicrobial proteins/peptides.
101
How does fever contribute to innate immunity?
It raises body temperature to inhibit pathogen growth and disrupt their metabolism.
102
How does a low pH environment help defend against pathogens?
The acidic conditions of the skin and stomach retard the growth of many microbes.
103
Name two key antimicrobial proteins found in innate immunity.
Lysozyme and lactoferrin.
104
What is the role of lysozyme?
It cleaves peptidoglycans in bacterial cell walls, particularly effective against gram-positive bacteria.
105
What does lactoferrin do in the immune response?
It sequesters essential nutrients like iron, inhibiting bacterial and fungal growth, and activates macrophages.
106
What are antimicrobial peptides?
Small, cationic peptides (e.g., defensins, cathelicidins) that disrupt microbial membranes.
107
What role do surfactant proteins play?
Found mainly in the respiratory tract, they bind bacterial surfaces and help clear pathogens while lubricating tissues.
108
What is pinocytosis?
A non-specific process where cells “drink” extracellular fluid through invagination of the plasma membrane.
109
What is receptor-mediated endocytosis?
A specific uptake process where receptors cluster in clathrin-coated pits to internalize targeted molecules.
110
What is phagocytosis?
A process by which specialized cells engulf and digest large particulate matter, such as pathogens.
111
Which cells are primarily responsible for phagocytosis?
Neutrophils and macrophages are the main phagocytes, with dendritic cells also contributing.
112
What are the four stages of phagocytosis?
Recognition, ingestion, digestion, and exocytosis (which may include antigen presentation).
113
How do phagocytes recognize pathogens directly?
Via pattern recognition receptors (PRRs) that bind to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs).
114
What is indirect recognition in phagocytosis?
It occurs when opsonins, such as antibodies or complement proteins, coat the pathogen for enhanced uptake.
115
What are opsonins?
Molecules (e.g., antibodies, complement proteins) that coat pathogens to facilitate recognition and phagocytosis.
116
What are Toll-like receptors (TLRs)?
A family of pattern recognition receptors that detect PAMPs and initiate inflammatory signaling pathways.
117
Which Toll-like receptor is known for recognizing LPS?
TLR4 recognizes lipopolysaccharide from gram-negative bacteria.
118
What signaling cascades are activated upon TLR engagement?
TLRs activate adaptor molecules (e.g., MyD88), triggering NF-κB and MAP kinase pathways that induce inflammatory mediators.
119
What is the role of intracellular PRRs?
They detect viral or bacterial nucleic acids (e.g., dsRNA, CpG DNA) and induce type I interferons and other cytokines.
120
Name two other classes of PRRs besides TLRs.
C-type lectin receptors (e.g., mannose receptor, dectin-1) and scavenger receptors.
121
What is the function of the mannose receptor on phagocytes?
It binds sugars such as mannose and fucose on pathogens, aiding in recognition and phagocytosis.
122
How do Fc receptors contribute to phagocytosis?
They bind the Fc portion of antibodies that coat pathogens, enhancing opsonization and uptake by phagocytes.
123
What role do complement receptors play in innate immunity?
They recognize complement proteins (e.g., C3b) deposited on pathogens, promoting phagocytosis.
124
What is the complement system’s function in innate immunity?
It opsonizes pathogens, forms the membrane attack complex to lyse cells, and recruits immune cells via chemotactic signals.
125
What is the significance of C-reactive protein (CRP)?
CRP is an acute-phase protein that binds pathogens and activates the classical complement pathway.
126
How do defensins kill bacteria?
They bind to and disrupt bacterial membranes by forming pores, leading to cell death.
127
What is the role of cathelicidins?
Cathelicidins disrupt microbial membranes, particularly on mucosal surfaces, aiding in pathogen clearance.
128
What are danger-associated molecular patterns (DAMPs)?
Molecules released from damaged or dying cells that signal tissue injury and trigger inflammation.
129
How do PRRs distinguish self from non-self?
They recognize conserved molecular structures (PAMPs) that are absent in host cells, while DAMPs indicate cell damage.
130
What is receptor clustering in phagocytosis?
The gathering of receptors at the cell surface to form a phagocytic cup, facilitating efficient pathogen uptake.
131
How does endocytosis differ from phagocytosis?
Endocytosis (including pinocytosis and receptor-mediated) is for smaller particles or fluid uptake, while phagocytosis is for large particulate matter.
132
Why is phagocytosis important for antigen presentation?
It allows antigen-presenting cells (like dendritic cells) to process pathogens and display fragments on MHC molecules to activate T cells.
133
How do innate and adaptive immune systems interact?
Innate responses activate and shape adaptive immunity through antigen presentation and cytokine release.
134
What role does NF-κB play in innate immunity?
NF-κB is a transcription factor that, once activated, induces the production of inflammatory cytokines and mediators.
135
What are MAP kinases, and why are they important?
MAP kinases relay signals from activated receptors to the nucleus, promoting the transcription of inflammatory genes.
136
How do antimicrobial peptides and proteins work together?
They disrupt microbial membranes, sequester nutrients, and activate immune cells—often acting synergistically (e.g., lysozyme with lactoferrin).
137
What is receptor-mediated endocytosis’s role in immune surveillance?
It allows cells to specifically internalize pathogens or antigens for enhanced processing and presentation.
138
Why is the innate immune response described as “ready from birth”?
Its components (physical barriers, phagocytes, PRRs) are constitutively expressed and require no prior exposure to function.
139
How do inflammatory responses contribute to pathogen clearance?
They recruit immune cells, increase vascular permeability, and produce cytokines/chemokines that coordinate pathogen elimination and tissue repair.
140
What is the role of natural killer (NK) cells in innate immunity?
NK cells are cytotoxic lymphocytes that kill virus-infected and tumor cells without prior sensitization.
141
How do innate lymphoid cells (ILCs) differ from NK cells?
ILCs are a diverse group that includes cells with helper functions, contributing to tissue homeostasis and inflammation.
142
How do NKT cells bridge innate and adaptive immunity?
NKT cells express T cell receptors yet recognize lipid antigens presented by CD1d, rapidly producing cytokines that influence both innate and adaptive responses.
143
What role do Interferon Regulatory Factors (IRFs) play in innate immunity?
IRFs are transcription factors activated (via TLR signaling) that induce type I interferon production, essential for antiviral responses.
144
Which adaptor protein is most commonly involved in TLR signaling?
MyD88 is the primary adaptor protein that transmits signals from most TLRs to activate downstream inflammatory pathways.
145
What is the significance of receptor clustering during phagocytosis?
Clustering of receptors at the cell surface forms a phagocytic cup, enhancing the efficiency of pathogen internalization.
146
Which complement receptors are primarily involved in phagocytosis?
Complement receptors CR1, CR3, and CR4 on phagocytes recognize complement-opsonized pathogens and mediate their uptake.
147
How does phagocytosis differ from pinocytosis?
Phagocytosis ingests large particles (>1 µm) by specialized cells, whereas pinocytosis involves non-specific uptake of extracellular fluid and solutes.
148
What are Danger-Associated Molecular Patterns (DAMPs)?
DAMPs are endogenous molecules released from damaged or dying cells that signal tissue injury and trigger inflammatory responses.
149
How do surface pattern recognition receptors (PRRs) differ from intracellular PRRs?
Surface PRRs (e.g., TLRs on the cell membrane) detect extracellular pathogens, while intracellular PRRs (e.g., RIG-I-like and NOD-like receptors) detect pathogens that invade the cytoplasm.
150
How do cytokines coordinate the innate immune response?
Cytokines recruit and activate immune cells, induce fever, and help amplify and direct the inflammatory response.
151
How does receptor-mediated endocytosis enhance antigen presentation?
It selectively internalizes antigens via specific receptors, allowing cells to process these antigens and present fragments on MHC molecules to T cells.
152
What happens during the ingestion stage of phagocytosis?
Receptor clustering triggers the formation of pseudopodia that surround and engulf the pathogen, forming a membrane-bound phagosome; this process requires energy and cytoskeletal rearrangement.
153
What are pseudopodia?
Projections of the cell membrane that extend around a pathogen to engulf it.
154
How does the endoplasmic reticulum contribute to phagocytosis?
The ER provides membranes used for forming phagosomes and pseudopodia, contributing to membrane recycling.
155
What occurs during the digestion stage in phagocytosis?
The phagosome fuses with lysosomes to form a phagolysosome, where enzymes degrade the engulfed pathogen.
156
Name two oxygen-independent antimicrobial mechanisms in the phagolysosome.
Acidification of the phagolysosome and the action of enzymes like lysozyme, defensins, lactoferrin, cathelicidins, and S100 proteins.
157
Name two oxygen-dependent mechanisms used in the phagolysosome.
The production of reactive oxygen intermediates (ROIs) and reactive nitrogen intermediates (RNIs) during the respiratory burst.
158
What is the respiratory burst?
A rapid increase in oxygen consumption by activated phagocytes that generates ROIs to kill pathogens.
159
Which enzyme is crucial for generating reactive oxygen intermediates?
NADPH oxidase, which converts oxygen into superoxide radicals.
160
How are reactive nitrogen intermediates produced?
Through inducible nitric oxide synthase (iNOS) converting arginine to nitric oxide (NO), which can combine with superoxide to form peroxynitrite.
161
What is the purpose of exocytosis in phagocytosis?
To release undigested debris, recycle membrane components, and present processed antigens for adaptive immune activation.
162
How does antigen presentation link innate and adaptive immunity?
Digested antigen fragments are loaded onto MHC molecules for presentation to T cells, triggering the adaptive immune response.
163
What are the four cardinal signs of inflammation?
Redness, swelling, heat, pain, and additionally a loss of function.
164
What is the acute phase response?
An early inflammatory response (0–24 hours) that increases blood flow, vascular permeability, and recruitment of leukocytes to the affected tissue.
165
Which cell type is central to initiating inflammation via mediator release?
Mast cells.
166
Which cell type is central to initiating inflammation via mediator release?
Mast cells, which release histamine to increase vascular permeability.
167
How do endothelial cells facilitate leukocyte extravasation during inflammation?
By upregulating adhesion molecules that enable rolling, firm adhesion, and transendothelial migration of leukocytes.
168
What are the main stages of leukocyte extravasation?
Rolling along the endothelium, firm adhesion, and transendothelial migration into the tissue.
169
What are cytokines?
Protein mediators produced by immune cells that regulate and coordinate the inflammatory response.
170
Name three pro-inflammatory cytokines involved in the acute phase response.
Interleukin-1 (IL-1), IL-6, and tumor necrosis factor-alpha (TNF-α).
171
Which cytokines serve as anti-inflammatory mediators?
Interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β).
172
What are chemokines?
Small proteins that function as chemoattractants to direct leukocyte migration along a concentration gradient.
173
What distinguishes CC and CXC chemokines?
They are two major chemokine families distinguished by the spacing of cysteine residues, which correlates with their specific roles in attracting different cell types.
174
Through what type of receptors do chemokines signal?
G protein-coupled receptors (GPCRs).
175
What are eicosanoids, and how do they affect inflammation?
Eicosanoids (prostaglandins, leukotrienes, thromboxanes) are derived from arachidonic acid and regulate vascular permeability and leukocyte migration.
176
Which enzyme is targeted by anti-inflammatory drugs like aspirin?
Cyclooxygenase (COX), particularly COX-2 in inflamed tissues.
177
How does histamine contribute to the inflammatory response?
Histamine increases vascular permeability and causes smooth muscle contraction, leading to redness and swelling.
178
What are neutrophil extracellular traps (NETs)?
Structures formed when neutrophils release chromatin and granule proteins to trap and kill pathogens extracellularly.
179
What is pyroptosis?
A form of programmed, inflammatory cell death initiated by inflammasome activation (e.g., NLRP3) that releases cytokines like IL-1β.
180
Which cytokine is closely associated with inflammasome activation and pyroptosis?
Interleukin-1β (IL-1β).
181
How do inflammatory mediators produce both local and systemic effects?
They enhance local blood flow, increase vascular permeability, recruit immune cells, and signal the hypothalamus to induce fever.
182
What role does the complement system play in inflammation?
It opsonizes pathogens, induces chemotaxis, and forms the membrane attack complex to lyse pathogens.
183
How does receptor-mediated endocytosis enhance pathogen uptake?
By internalizing opsonized pathogens via specific receptors (e.g., Fc receptors, complement receptors) to facilitate efficient phagocytosis.
184
Why is cytoskeletal rearrangement important during phagocytosis?
It enables the formation of pseudopodia and the engulfment of pathogens, making phagocytosis an energy-dependent process.
185
How does energy metabolism impact phagocytosis?
Adequate ATP is required for actin polymerization and other energy-dependent processes essential for effective phagocytosis.
186
What are the differences between macrophages and neutrophils in antimicrobial activity?
Neutrophils produce a higher respiratory burst and abundant defensins for rapid killing, while macrophages use sustained lysosomal degradation and enzyme production.
187
How do phagocytes use receptor-mediated endocytosis to enhance antigen presentation?
They selectively internalize opsonized pathogens, process them, and present antigen fragments on MHC molecules to activate T cells.
188
What is the significance of linking phagocytosis with antigen presentation?
It bridges innate and adaptive immunity, enabling a tailored T cell response to the ingested pathogen.
189
What is the difference between direct and indirect recognition during phagocytosis?
Direct recognition occurs when phagocytes use pattern recognition receptors (PRRs) to bind pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs); indirect recognition happens when opsonins (such as antibodies or complement proteins) coat pathogens, allowing receptors like Fc or complement receptors to mediate uptake.
190
What role does the endoplasmic reticulum (ER) play in phagocytosis?
The ER provides membranes that are used to form pseudopodia and phagosomes, and it assists in recycling membranes during endocytosis.
191
How do the antigen presentation pathways differ for MHC class I and class II in relation to phagocytosis?
MHC class I presents peptides derived from cytosolic degradation (typical for intracellular pathogens), whereas MHC class II presents peptides from phagolysosomal digestion of ingested pathogens, linking innate and adaptive immunity.
192
Why is receptor clustering important in phagocytosis?
Receptor clustering triggers cytoskeletal rearrangements that lead to pseudopodia formation and efficient engulfment of the pathogen.
193
How do chemokines mediate leukocyte migration during inflammation?
Chemokines bind to G protein–coupled receptors (GPCRs) on leukocytes, activating signaling pathways that reorganize the cytoskeleton and direct cells along a concentration gradient toward the site of infection or injury.
194
How can serum manipulation be used experimentally to study phagocytosis?
By comparing phagocytosis with and without serum—or using serum deficient in specific complement components—researchers can distinguish between direct receptor-mediated uptake and opsonin-enhanced (indirect) phagocytosis.
195
What experimental evidence supports the role of receptor clustering in phagocytosis?
Knockout studies and serum manipulation experiments show that when receptors (such as Fc or complement receptors) are absent or not clustered, the efficiency of pseudopodia formation and subsequent pathogen uptake is significantly reduced.
196
What are the four interconnected enzyme cascades that contribute to inflammation?
The kinin, clotting, fibrinolytic, and complement cascades.
197
How is the kinin system activated and what mediator does it produce?
Tissue injury activates Factor XII, which converts pre‑kallikrein to kallikrein; kallikrein then cleaves kininogen to form bradykinin, which increases vascular permeability, causes vasodilation, pain, and smooth muscle contraction.
198
What role does the clotting system play in inflammation?
Activation of Factor XII leads to thrombin generation, which converts fibrinogen to fibrin, forming clots. Fibrinopeptides released during clot formation increase vascular permeability and attract neutrophils.
199
What is the function of the fibrinolytic system in the context of inflammation?
It is triggered by endothelial damage, leading to plasmin production that degrades fibrin clots into chemotactic fragments and contributes to complement activation.
200
What are the three pathways of complement activation?
The classical pathway, lectin pathway, and alternative pathway.
201
How is the classical complement pathway initiated?
By binding of antibodies (IgG or IgM) or CRP to a pathogen, which recruits the C1 complex that cleaves C4 and C2 to form the C3 convertase.
202
What is the role of C3 convertase in the complement cascade?
It cleaves C3 into C3a and C3b; C3b opsonizes pathogens and, together with other components, forms C5 convertase.
203
How does the lectin pathway differ from the classical pathway?
The lectin pathway is activated by mannose‑binding lectin (MBL) binding to sugars on the pathogen surface, which then recruits MASPs to cleave complement components similarly to the classical pathway.
204
Describe how the alternative complement pathway is activated.
It involves the spontaneous cleavage of C3 into C3a and C3b; C3b binds directly to microbial surfaces, associates with Factor B and D (stabilized by properdin) to form an alternative C3 convertase.
205
What are the primary outcomes of complement activation?
Opsonization of pathogens, generation of anaphylatoxins (C3a, C5a) that mediate inflammation and chemotaxis, and formation of the membrane attack complex (MAC) that causes cell lysis.
206
How is the membrane attack complex (MAC) formed and what does it do?
Sequential binding of complement components C5b, C6, C7, C8, and multiple C9 molecules forms the MAC, which inserts into pathogen membranes to cause lysis.
207
What roles do C3a and C5a play in the inflammatory response?
They act as anaphylatoxins that promote chemotaxis, activate phagocytes, and trigger the respiratory burst while increasing vascular permeability.
208
What is cell-mediated innate immunity?
It encompasses the rapid cytotoxic responses by innate lymphoid cells—such as NK cells, NKT cells, γδ T cells, and B1 cells—that kill infected or abnormal cells without prior sensitization.
209
What are the three main mechanisms by which natural killer (NK) cells kill target cells?
NK cells kill via (1) perforin/granzyme-mediated cytotoxicity, (2) Fas ligand-induced apoptosis, and (3) antibody-dependent cellular cytotoxicity (ADCC) through CD16.
210
How do natural killer T (NKT) cells bridge innate and adaptive immunity?
NKT cells express T cell receptors that recognize lipid antigens presented on CD1 molecules, rapidly secreting cytokines that influence both innate and adaptive responses.
211
What characterizes γδ T cells in innate immunity?
They are predominantly located in mucosal tissues and the skin, recognize non-peptide antigens, and can directly kill infected cells and secrete cytokines.
212
What is the role of B1 cells in innate immunity?
B1 cells produce natural, low-affinity IgM antibodies against common bacterial carbohydrate antigens, playing a critical role in early defense, especially in neonates.
213
How do the clotting, kinin, fibrinolytic, and complement systems interact during inflammation?
They are interlinked cascades—initiated by tissue injury—that amplify inflammation, coordinate immune cell recruitment, and work together to contain pathogens and promote tissue repair.
214
What is the significance of Factor XII in these enzyme cascades?
Factor XII, when activated by tissue injury, initiates the kinin, clotting, and fibrinolytic cascades, linking vascular injury to inflammatory and hemostatic responses.
215
How do cytokines and chemokines regulate the inflammatory response?
Cytokines (e.g., IL-1, TNF-α, IL-6) initiate and amplify inflammation, while chemokines guide the migration of leukocytes to sites of injury via GPCR signaling.
216
What role do GPCRs play in leukocyte chemotaxis?
GPCRs on leukocytes bind chemokines, triggering intracellular signaling that reorganizes the cytoskeleton and directs cell migration along a concentration gradient.
217
How do mast cells contribute to the inflammatory cascade?
Mast cells release histamine and other mediators that increase vascular permeability and promote leukocyte adhesion and extravasation.
218
What are acute-phase proteins and how do they function in inflammation?
Acute-phase proteins (such as CRP, serum amyloid A, and MBL) are produced by the liver in response to cytokines and act to opsonize pathogens and modulate the complement cascade.
219
How do proteases function within the inflammatory cascades?
Proteases from the kinin, clotting, and fibrinolytic systems generate active mediators (e.g., bradykinin, thrombin, plasmin) that increase vascular permeability, promote chemotaxis, and assist in clot formation and degradation.
220
What effects does bradykinin have during inflammation?
Bradykinin causes vasodilation, increases vascular permeability, induces pain, and promotes smooth muscle contraction.
221
What is the role of thrombin in the clotting cascade and inflammation?
Thrombin converts fibrinogen into fibrin, forming clots and releasing fibrinopeptides that enhance vascular permeability and neutrophil chemotaxis.
222
How does the fibrinolytic system resolve clots, and what additional role does it play?
Plasmin degrades fibrin clots into chemotactic fragments and helps clear clots, further modulating the inflammatory response and aiding complement activation.
223
How do complement fragments like C3a and C5a enhance the inflammatory response?
They function as potent chemotactic agents, stimulate the respiratory burst in phagocytes, and trigger degranulation of mast cells.
224
What experimental methods can distinguish between direct and indirect phagocytosis?
Comparing phagocytosis with serum-free conditions (direct recognition) versus using normal or complement-deficient serum (indirect opsonization) reveals the contribution of opsonins.
225
What is the role of receptor clustering in phagocytosis?
Clustering of receptors facilitates the formation of pseudopodia and the engulfment of pathogens, making the process energy dependent and efficient.
226
How does receptor-mediated endocytosis aid antigen presentation?
It selectively internalizes opsonized pathogens, allowing for processing and subsequent presentation of antigen fragments on MHC molecules to T cells.
227
How do the complement pathways (classical, lectin, alternative) converge?
All three pathways lead to the activation of C3, resulting in opsonization, generation of anaphylatoxins, and formation of the membrane attack complex (MAC).
228
How does the interplay of complement activation and inflammatory mediators affect tissue damage?
While essential for pathogen clearance, excessive complement activation and mediator release can lead to collateral tissue damage due to cytotoxic effects and oxidative stress.
229
How do natural killer (NK) cells decide to kill a target cell?
NK cells use a balance of activatory and inhibitory receptors to detect cells with reduced MHC class I expression or stress-induced ligands, triggering cytotoxicity.
230
What is antibody-dependent cellular cytotoxicity (ADCC) and which cell mediates it?
ADCC is a process where NK cells recognize antibodies bound to target cells through CD16, leading to the release of cytotoxic granules and target cell death.
231
How do CD1 molecules function in innate immunity?
CD1 molecules present lipid antigens to NKT cells, allowing for recognition and rapid cytokine production that bridges innate and adaptive responses.
232
What is the overall significance of cell-mediated innate immunity?
It provides rapid cytotoxic responses against infected or abnormal cells, supplements phagocytosis, and supports the initiation of adaptive immune responses through cytokine secretion.
233
How does the timing of cytokine production regulate the inflammatory response?
Early release of pro‑inflammatory cytokines (e.g., IL‑1, TNF‑α) initiates inflammation and recruits immune cells; later, anti‑inflammatory cytokines (e.g., IL‑10, TGF‑β) help resolve inflammation and promote tissue repair.
234
What role do macrophages play in handling heavy metals and degradation products?
After phagocytosis, macrophages can store heavy metals (like iron) and other remnants, which may influence subsequent tissue responses and repair.
235
What is autophagy and how does it support innate immunity?
Autophagy is a cellular process that degrades and recycles intracellular components—including pathogens—thereby reducing infection and enhancing antigen presentation.
236
How is peroxynitrite formed and what is its function?
Peroxynitrite is generated when nitric oxide reacts with superoxide; it is a potent oxidant that damages microbial components, enhancing pathogen killing.
237
How can serum manipulation experiments help distinguish between direct and indirect phagocytosis?
By comparing phagocytosis under serum‑free conditions versus serum with normal or deficient complement levels, researchers can assess the role of opsonins (antibodies/complement) in enhancing pathogen uptake.
238
How do complement fragments like C3a and C5a function as chemoattractants?
C3a and C5a bind to specific receptors on immune cells, inducing chemotaxis and promoting the recruitment of phagocytes to infection sites.
239
What is an antigen?
An antigen is any substance that binds to specific receptors on lymphocytes, triggering a specific immune response.
240
How does an immunogen differ from an antigen?
An immunogen is an antigen that elicits an immune response; while all immunogens are antigens, not every antigen is immunogenic.
241
What is an epitope (antigenic determinant)?
An epitope is the specific region or fragment of an antigen that is recognized and bound by an antigen receptor on a lymphocyte.
242
What is a hapten?
A hapten is a small molecule that can act as an epitope but, by itself, does not elicit an immune response unless attached to a larger carrier protein.
243
What types of molecules can serve as antigens?
Antigens may be proteins, lipids, carbohydrates, or any combination of these, and they can be either foreign or altered self molecules.
244
How can antigens enter the body?
Antigens enter through breaches in skin or mucosal membranes, direct injection (e.g., bites, needles), organ transplants, skin grafts, or via specialized M cells in mucosal surfaces.
245
What is the difference between simple and complex antigens?
Simple antigens are small, single molecules (like ovalbumin or pollen), while complex antigens consist of multiple antigenic determinants (such as bacterial cell wall components) on a larger structure.
246
What is the role of adjuvants in vaccination?
Adjuvants enhance the immune response by increasing antigen persistence, effective size (for uptake by APCs), and by activating dendritic cells and macrophages to produce inflammatory cytokines.
247
Which type of antigen is recognized directly by B cell receptors?
B cell receptors recognize native, three-dimensional conformations of antigens that are free in solution or suspension.
248
How do T cell receptors (TCRs) recognize antigens differently than B cell receptors?
TCRs recognize short, linear peptide fragments (typically 9–20 amino acids) that are processed and presented on MHC molecules by antigen-presenting cells.
249
What is the structure of a typical immunoglobulin molecule?
It is a flexible Y-shaped molecule composed of two identical heavy chains and two identical light chains joined by disulfide bonds, with variable regions at the tips forming the antigen-binding sites.
250
What are the complementarity-determining regions (CDRs)?
CDRs are hypervariable loops in the variable regions of immunoglobulins that determine the specificity and affinity of antigen binding.
251
How many classes of immunoglobulins are there and what are they?
There are five classes: IgM, IgG, IgA, IgD, and IgE.
252
Which immunoglobulin class is primarily found on the surface of naïve B cells?
Naïve B cells predominantly express IgM (and IgD) as their B cell receptor.
253
What happens during class-switch recombination in B cells?
Class-switch recombination changes the constant region of the immunoglobulin heavy chain, allowing the B cell to produce antibodies of a different class (e.g., IgG, IgA, or IgE) while retaining the same antigen specificity.
254
What is the B cell receptor (BCR) complex?
The BCR complex consists of the membrane-bound immunoglobulin (mIg) along with accessory molecules (Igα and Igβ, also known as CD79α and CD79β) that contain ITAMs for intracellular signaling.
255
What is the constant region of the immunoglobulin heavy chain?
The constant region allows the B cell to produce antibodies of a different class (e.g., IgG, IgA, or IgE) while retaining the same antigen specificity.
256
Why is the ITAM important in the BCR complex?
ITAMs (immunoreceptor tyrosine-based activation motifs) in the Igα and Igβ chains transmit signals into the cell upon antigen binding, leading to B cell activation and subsequent clonal expansion.
257
What is the relationship between the B cell receptor and secreted antibodies?
The secreted antibody and the B cell receptor have identical variable regions and antigen specificity; the BCR is the membrane-bound form of the antibody.
258
How does the structure of an antibody determine its effector functions?
The constant region of the antibody, which differs among classes, mediates effector functions such as complement activation, binding to Fc receptors on cells, and determining antibody distribution.
259
What structural difference exists between membrane-bound BCRs and secreted antibodies?
Membrane-bound BCRs contain a transmembrane domain and a short cytoplasmic tail, whereas secreted antibodies lack the transmembrane domain and are released into circulation.
260
What is the significance of the heavy chain constant regions (CH domains) in antibodies?
The CH domains (e.g., CH1, CH2, CH3) determine the class of the antibody and dictate its effector functions and interactions with complement and Fc receptors.
261
What are the two types of light chains in immunoglobulins?
The two types are kappa (κ) and lambda (λ) light chains.
262
How does the diversity of the B cell receptor repertoire arise?
Diversity is generated through random gene rearrangement of variable (V), diversity (D), and joining (J) gene segments during B cell development.
263
What role does antigen binding play in B cell activation?
Binding of antigen to the BCR triggers intracellular signaling via ITAMs, leading to B cell clonal expansion, differentiation into plasma cells, and formation of memory B cells.
264
How does the immune system achieve specificity in the adaptive immune response?
Specificity is achieved by the unique antigen-binding sites on BCRs and TCRs, generated through gene rearrangements that produce a diverse repertoire capable of recognizing millions of different epitopes.
265
What is the central principle behind vaccine action?
Vaccines work by introducing an antigen (often with an adjuvant) that elicits a specific immune response and generates long-lasting immunological memory without causing disease.
266
How are antigens processed for T cell recognition?
Antigens are internalized, processed into peptide fragments, and presented on MHC molecules by antigen-presenting cells for T cell recognition.
267
Why might an antigen fail to elicit an immune response?
If the antigen is too small (a hapten), not in its native structure, or similar to self molecules, it may not be immunogenic without being attached to a carrier or without proper adjuvant support.
268
What factors influence an antigen's immunogenicity?
Factors include molecular size, complexity, dose, route of entry, and similarity to self-proteins.
269
How do adjuvants enhance the immunogenicity of an antigen?
Adjuvants increase antigen persistence, effective size, and activate innate immune cells, thereby promoting stronger antigen uptake and presentation.
270
What is an epitope?
An epitope, or antigenic determinant, is the specific part of an antigen that is recognized and bound by an antibody or T cell receptor.
271
What is the difference between a linear and a discontinuous epitope?
A linear epitope consists of a continuous sequence of amino acids, whereas a discontinuous epitope is formed by amino acids that are separated in the primary sequence but come together in the protein’s 3D structure.
272
Define affinity in the context of antigen–antibody interactions.
Affinity is the strength of the binding interaction between a single antigen-binding site of an antibody and a single epitope.
273
How does avidity differ from affinity?
Avidity is the overall strength of binding between an antibody (with multiple binding sites) and an antigen with multiple epitopes; it represents the combined strength of all individual interactions.
274
Why can IgM antibodies be effective despite having low individual binding affinity?
IgM antibodies form pentamers, giving them ten antigen-binding sites that collectively provide high avidity, compensating for their lower individual affinity.
275
What are the two main types of antigen receptors in adaptive immunity?
B cell receptors (BCRs) and T cell receptors (TCRs).
276
How do BCRs recognize antigen compared to TCRs?
BCRs bind to native antigens in their three-dimensional conformation, while TCRs recognize processed peptide fragments presented on MHC molecules.
277
What structural components make up an antibody molecule?
An antibody consists of two identical heavy chains and two identical light chains, with variable regions (for antigen binding) and constant regions (for effector functions) joined by disulfide bonds.
278
What are Complementarity Determining Regions (CDRs)?
CDRs are hypervariable loops within the variable regions of immunoglobulin heavy and light chains that directly interact with the epitope and determine specificity.
279
How does the B cell receptor (BCR) relate to secreted antibodies?
The BCR is the membrane-bound form of the antibody; they share identical variable regions and antigen specificity, differing mainly by the presence (BCR) or absence (secreted antibody) of a transmembrane domain.
280
What are the five classes of antibodies?
IgM, IgG, IgA, IgD, and IgE.
281
Which antibody classes are typically produced after B cell activation and class switching?
IgG, IgA, and IgE are produced after class switching, while naïve B cells primarily express IgM and IgD.
282
What is the B cell receptor complex?
It comprises the membrane-bound immunoglobulin (BCR) along with the Igα and Igβ (CD79α/CD79β) heterodimers that contain ITAMs for signal transduction.
283
Why do BCRs require accessory molecules like Igα and Igβ?
The BCR’s cytoplasmic tail is too short to signal; Igα and Igβ contain ITAMs that initiate intracellular signaling upon antigen binding.
284
What is meant by 'polyclonal response' in an immune reaction?
A polyclonal response involves the activation and expansion of multiple clones of B or T cells, each recognizing different epitopes of the same antigen or different antigens on a pathogen.
285
How does a monoclonal antibody differ from a polyclonal response?
Monoclonal antibodies are produced by a single B cell clone and recognize one specific epitope, whereas polyclonal responses involve antibodies from multiple clones recognizing various epitopes.
286
What is the structure of a T cell receptor (TCR)?
TCRs are heterodimers (usually composed of α and β chains, or less commonly γ and δ chains) with variable regions for antigen binding and constant regions; they remain membrane-bound.
287
How do TCRs recognize antigens?
TCRs recognize short peptide fragments (typically 9–20 amino acids) that are processed and presented by MHC molecules on antigen-presenting cells.
288
What accessory molecules are part of the TCR complex, and what is their role?
The TCR complex includes CD3 molecules (γ, δ, ε, and ζ chains) that contain ITAMs for signal transduction and co-receptors (CD4 or CD8) that stabilize interactions with MHC molecules.
289
How does the antigen-binding specificity of TCRs compare to that of BCRs?
TCRs are specific for peptide–MHC complexes and have a single antigen-binding site per TCR, whereas BCRs bind native antigen in their native conformation and typically have two binding sites per molecule.
290
What is the significance of MHC restriction in T cell activation?
T cells can only recognize antigens when they are presented on MHC molecules, ensuring that T cell responses are targeted and self-tolerance is maintained.
291
What are superantigens, and how do they affect T cell activation?
Superantigens are microbial proteins that bypass normal antigen processing by binding non-specifically to the variable region of TCRs and non-polymorphic regions of MHC class II, activating a large proportion (5–20%) of T cells.
292
What is the difference between αβ and γδ T cells?
αβ T cells, which are the majority in circulation, recognize peptides presented by MHC molecules, while γδ T cells are less diverse, are enriched in epithelial tissues, and can recognize non-peptide antigens (often presented by CD1 molecules).
293
How do TCRs and BCRs share structural similarities?
Both contain variable and constant regions built from immunoglobulin folds, and both rely on gene rearrangement to generate diversity in their antigen-binding sites.
294
What are the paratopes in antigen receptors?
Paratopes are the antigen-binding sites on antibodies or T cell receptors, composed of the variable regions that interact with the epitope.
295
How do antigen receptors contribute to the specificity of the adaptive immune response?
Their unique variable regions, generated by gene rearrangement, allow each lymphocyte to recognize a specific epitope, enabling a highly targeted immune response.
296
What factors influence the immunogenicity of an antigen?
Factors include molecular size, complexity, dose, route of entry, and similarity to self-antigens.
297
How do adjuvants enhance the immunogenicity of an antigen?
Adjuvants increase antigen persistence, effective size (for uptake by APCs), and stimulate innate immune responses, thereby promoting stronger adaptive responses.
298
What is meant by 'affinity maturation' in B cells?
Affinity maturation is the process by which B cells undergo somatic hypermutation in germinal centers to increase the binding strength (affinity) of their antibodies for the antigen.
299
How do the variable regions of antibodies contribute to antigen diversity?
The variable regions are generated by random gene rearrangements and somatic mutations, allowing the immune system to produce a vast repertoire of antibodies with diverse specificities.
300
What are the roles of the constant regions in antibodies?
Constant regions determine the class of the antibody and mediate effector functions such as complement activation and binding to Fc receptors on immune cells.
301
What is class-switch recombination?
It is the process by which activated B cells change the constant region of their immunoglobulin heavy chain, resulting in the production of different antibody isotypes (e.g., switching from IgM to IgG) while retaining antigen specificity.
302
How do T cells become activated upon antigen recognition?
T cell activation requires the binding of the TCR to a peptide–MHC complex, stabilization by co-receptors (CD4 or CD8), and signal transduction via the CD3 complex and associated kinases.
303
What is the role of the immunological synapse?
The immunological synapse is the interface between a T cell and an antigen-presenting cell where TCR, co-receptors, and adhesion molecules organize to facilitate effective signaling and activation.
304
What is the significance of polyclonal responses during an infection?
A polyclonal response involves multiple B and T cell clones recognizing various epitopes of a pathogen, providing a broad and robust defense against complex antigens.
305
Why are monoclonal antibodies useful in research and therapy?
They are derived from a single B cell clone and recognize one specific epitope, offering consistent specificity and high affinity for diagnostic or therapeutic purposes.
306
How does the adaptive immune system generate memory?
After clonal expansion, a small population of activated lymphocytes differentiates into long-lived memory cells that enable a faster, more robust response upon re-exposure to the same antigen.
307
What is the role of antigen processing in T cell activation?
Antigen-presenting cells internalize antigens, process them into peptide fragments, and present these fragments on MHC molecules, which are then recognized by TCRs.
308
How do co-receptors (CD4 and CD8) assist in T cell activation?
Co-receptors bind to conserved regions of MHC molecules, stabilizing the TCR–peptide–MHC interaction and enhancing signal transduction.
309
What is the overall significance of antigen–antigen receptor interactions?
These interactions confer specificity to the adaptive immune response, ensuring that B and T cells precisely recognize and eliminate pathogens while also generating immunological memory.
310
What are superantigens and what effect do they have on T cell populations?
Superantigens are microbial proteins that bind non-specifically to conserved regions of TCRs and MHC class II molecules, bypassing normal antigen processing and activating up to 5–20% of T cells, which can lead to an overwhelming cytokine release (cytokine storm).
311
How do T cells discriminate between self and non-self antigens?
T cells undergo positive and negative selection in the thymus; those that bind self-peptides too strongly are eliminated (negative selection), while only cells that can interact with MHC survive (positive selection), ensuring self-tolerance.
312
What key signaling motifs are found in the TCR complex and what is their function?
The CD3 complex contains ITAMs (immunoreceptor tyrosine-based activation motifs) that, upon TCR engagement, become phosphorylated to initiate intracellular signaling cascades necessary for T cell activation.
313
Which enzyme is crucial for class-switch recombination in B cells, and what is its role?
Activation-induced cytidine deaminase (AID) initiates class-switch recombination by deaminating cytosine bases in switch regions, allowing B cells to recombine different constant region exons and change antibody isotype without altering antigen specificity.
314
What is the functional significance of a polyclonal response during an infection?
A polyclonal response involves multiple B and T cell clones recognizing different epitopes on a pathogen, ensuring a broad, robust, and effective immune defense against complex antigens.
315
What are the two main classes of MHC molecules?
MHC class I and MHC class II.
316
What is the basic structure of an MHC class I molecule?
It consists of a single α-chain with three domains (α1, α2, α3) that form a peptide-binding groove, and is non-covalently associated with β₂-microglobulin.
317
On which cells are MHC class I molecules expressed?
They are expressed on all nucleated cells (and platelets).
318
What is the role of β₂-microglobulin in MHC class I?
β₂-microglobulin stabilizes the MHC class I α-chain and is essential for proper folding and surface expression of the molecule.
319
What types of peptides does MHC class I typically present?
MHC class I presents endogenous peptides, typically 8–10 amino acids long, derived from intracellular proteins (e.g., viral proteins or mutated self-proteins).
320
Briefly describe the endogenous antigen processing pathway for MHC class I.
Intracellular proteins are ubiquitinated and degraded by the proteasome into peptides, which are then translocated into the endoplasmic reticulum (ER) by TAP, loaded onto MHC class I molecules, and transported to the cell surface.
321
What is the role of the proteasome in MHC class I antigen processing?
It degrades ubiquitinated proteins into peptide fragments suitable for loading onto MHC class I molecules.
322
What is TAP and what role does it play in MHC class I processing?
TAP (Transporter associated with Antigen Processing) translocates peptides from the cytosol into the ER, where they can be loaded onto MHC class I molecules.
323
How are MHC class I molecules stabilized before peptide binding?
They are maintained in a peptide-receptive state by chaperone proteins (e.g., calnexin, calreticulin) and the peptide-loading complex (including tapasin).
324
What is the basic structure of an MHC class II molecule?
MHC class II is composed of two polypeptide chains (α and β), each contributing two domains, forming a peptide-binding groove; both chains are transmembrane proteins.
325
Which cells predominantly express MHC class II molecules?
Professional antigen-presenting cells (APCs) such as dendritic cells, macrophages, and B cells.
326
What role does the invariant chain (Ii) play in MHC class II processing?
The invariant chain binds to the MHC class II peptide-binding groove in the ER, preventing premature peptide binding and directing the complex to the MIIC (MHC class II–enriched compartment).
327
What is CLIP and what is its function in MHC class II processing?
CLIP (Class II-associated Invariant chain Peptide) is the fragment of the invariant chain that remains in the peptide-binding groove until it is replaced by an antigenic peptide.
328
What role does HLA-DM play in MHC class II antigen presentation?
HLA-DM facilitates the removal of CLIP and the binding of antigenic peptides to MHC class II molecules.
329
What is the typical peptide length for antigens presented by MHC class II?
Peptides presented by MHC class II are usually 13–18 amino acids long.
330
How does peptide binding differ between MHC class I and class II molecules?
MHC class I binds shorter peptides (8–10 aa) from intracellular proteins, while MHC class II binds longer peptides (13–18 aa) derived from extracellular antigens processed in endosomes.
331
How do MHC class I and class II molecules differ in antigen presentation for T cell activation?
MHC class I presents endogenous peptides to CD8⁺ cytotoxic T cells, whereas MHC class II presents exogenous peptides to CD4⁺ helper T cells.
332
What is the overall purpose of antigen processing and presentation?
To display peptide fragments on the cell surface in the context of MHC molecules so that T cells can recognize and initiate a specific adaptive immune response.
333
What are the main pathways for antigen processing in MHC presentation?
The endogenous pathway for MHC class I and the exogenous pathway for MHC class II.
334
What is the HLA complex and where is it located?
The HLA complex (Human Leukocyte Antigen) is a set of genes on chromosome 6 that encode MHC class I and II molecules, along with other immune-related proteins.
335
Which genes encode MHC class I molecules in humans?
HLA-A, HLA-B, and HLA-C encode MHC class I molecules.
336
Which genes encode MHC class II molecules in humans?
HLA-DP, HLA-DQ, and HLA-DR encode MHC class II molecules.
337
What does it mean that MHC genes are highly polymorphic?
They exist in many different allelic forms, especially in the peptide-binding regions, allowing a diverse range of peptides to be presented within a population.
338
What is the significance of co-dominant expression of MHC genes?
Both alleles inherited from each parent are expressed, increasing the diversity of antigen presentation in an individual.
339
How does MHC diversity benefit a population?
It increases the likelihood that some individuals can present and respond to emerging pathogens, providing resilience against infections on a population level.
340
How does antigen processing differ between the endogenous and exogenous pathways?
The endogenous pathway (MHC I) processes intracellular proteins via the proteasome, while the exogenous pathway (MHC II) processes extracellular antigens in endocytic compartments.
341
What role do dendritic cells play in antigen presentation?
Dendritic cells capture antigens in peripheral tissues, process them, and migrate to lymph nodes where they present peptide–MHC complexes to T cells.
342
Why is peptide binding critical for the stability of MHC molecules on the cell surface?
Peptide binding stabilizes MHC molecules, preventing their rapid recycling and ensuring they remain on the cell surface for T cell surveillance.
343
Why do empty MHC molecules not persist on the cell surface?
Without bound peptides, MHC molecules are unstable and are rapidly internalized and degraded.
344
What is the immunological synapse?
It is the structured interface between a T cell and an antigen-presenting cell where TCR, co-receptors, and adhesion molecules aggregate to facilitate effective T cell activation.
345
What are the main differences in antigen source between MHC class I and II pathways?
MHC class I presents peptides derived from intracellular (endogenous) proteins, while MHC class II presents peptides from extracellular (exogenous) proteins.
346
How do the structural differences between MHC class I and II influence the peptides they bind?
MHC class I, with a closed-ended peptide-binding groove, binds short peptides (8–10 aa); MHC class II, with an open-ended groove, accommodates longer peptides (13–18 aa).
347
How does antigen presentation by MHC molecules bridge innate and adaptive immunity?
It allows APCs to display processed peptides to T cells, thus linking the detection of pathogens (innate immunity) with the activation of antigen-specific T cells (adaptive immunity).
348
What is the role of TAP in MHC class I processing?
TAP transports peptides from the cytosol into the ER where they are loaded onto MHC class I molecules.
349
What is the peptide loading complex in the context of MHC class I?
It is a group of chaperone proteins (e.g., tapasin, calreticulin, ERp57) that assist in peptide binding to MHC class I molecules, ensuring proper assembly and stability.
350
How are exogenous antigens processed for MHC class II presentation?
Antigens are internalized via endocytosis or phagocytosis, degraded in endosomes/lysosomes, and loaded onto MHC class II molecules within specialized compartments (MIIC).
351
What role does the invariant chain (Ii) play in MHC class II processing?
Ii binds to the MHC class II peptide-binding groove in the ER to block premature peptide binding and directs the MHC class II molecule to endosomal compartments.
352
How does HLA-DM facilitate antigen loading on MHC class II molecules?
HLA-DM promotes the release of CLIP (the remnant of Ii) from the MHC class II groove and stabilizes the binding of antigenic peptides.
353
What is the significance of MHC polymorphism in transplant rejection?
Because MHC molecules are highly polymorphic and co-dominantly expressed, mismatched MHC between donor and recipient can trigger strong immune rejection responses.
354
How does gene rearrangement contribute to the diversity of antigen receptors?
V(D)J recombination randomly rearranges gene segments in B and T cell receptors, generating a vast repertoire of receptors that can recognize millions of different antigens.
355
How many MHC alleles have been identified, and why is this significant?
Over 15,000 alleles have been identified, greatly enhancing the diversity of peptide presentation in the human population and increasing resilience to pathogens while complicating organ transplantation.
356
What is the role of ERAP in MHC class I antigen processing?
ERAP (Endoplasmic Reticulum Aminopeptidase) trims peptide fragments to the optimal 8–10 amino acids required for stable binding to MHC class I molecules.
357
What is cross-presentation in antigen processing?
Cross-presentation is a process where certain dendritic cells present exogenous antigens on MHC class I molecules, thereby activating CD8⁺ cytotoxic T cells.
358
What are professional antigen-presenting cells (APCs), and which cell types qualify?
Professional APCs are cells specialized in processing and presenting antigens via MHC class II; they include dendritic cells, macrophages, and B cells.
359
How do dendritic cells bridge innate and adaptive immunity?
They capture antigens in peripheral tissues, process them, and migrate to lymph nodes where they present peptide–MHC complexes to T cells, initiating the adaptive response.
360
What is the significance of the peptide binding groove’s structure in MHC molecules?
The groove’s structure determines the peptide length and specificity—MHC class I has a closed groove (binding 8–10 aa peptides), while MHC class II has an open groove (binding 13–18 aa peptides).
361
Why is peptide loading critical for MHC stability on the cell surface?
Peptide binding stabilizes MHC molecules; empty MHC molecules are unstable and quickly recycled, reducing the efficiency of antigen presentation.
362
How does the antigen presentation process differ between endogenous and exogenous pathways?
The endogenous pathway (MHC class I) processes intracellular proteins via the proteasome and TAP, while the exogenous pathway (MHC class II) involves uptake, degradation in endosomes, and peptide loading in MIIC compartments.
363
What role does the invariant chain (Ii) play in MHC class II processing?
The invariant chain blocks the peptide-binding groove in the ER to prevent premature peptide binding and directs the MHC class II molecule to endosomal compartments for peptide loading.
364
How do HLA-DM and HLA-DO regulate MHC class II peptide loading?
HLA-DM facilitates the release of CLIP and the binding of antigenic peptides, while HLA-DO modulates HLA-DM activity, fine-tuning the peptide repertoire loaded onto MHC class II.
365
Why is the co-dominant expression of MHC genes important?
It ensures that both parental alleles are expressed, maximizing the variety of peptides that can be presented and enhancing immune system diversity.
366
How does the stability of MHC-peptide complexes affect T cell surveillance?
Stable MHC-peptide complexes persist on the cell surface, increasing the chances that circulating T cells will encounter and recognize them, which is essential for effective immune monitoring.
367
What is the overall impact of antigen processing and presentation on adaptive immunity?
It is critical for activating T cells, shaping the specificity of the immune response, and ensuring long-lasting immunological memory against pathogens.
368
What is adaptive immunity?
An immune response that becomes more powerful following repeated encounters with the same antigen, using highly specific antigen receptors to recognise diverse pathogens and build immunological memory.
369
What are the three hallmarks of adaptive immunity?
1) Highly diverse cell populations, 2) Pathogen-specific clonal expansion, 3) Resolution (contraction) and memory formation.
370
How can specific adaptive immunity be acquired?
Naturally, via infection (e.g., chickenpox) or artificially, via vaccination (e.g., MMR, COVID).
371
Why do we need T cells?
To clear intracellular microbes in infected cells (CMI) and to help B cells produce varied Ig isotypes and high-affinity antibodies.
372
Where does T cell maturation and selection occur?
In the thymus, using positive selection for MHC recognition and negative selection to delete self-reactive cells (central tolerance).
373
Define naïve, effector, and memory T cells.
* Naïve: Never stimulated since thymic maturation. * Effector: Short-lived, activated, perform specialised functions (cytokine secretion, cytolysis). * Memory: Long-lived, resting state after activation, rapidly reactivated on re-exposure.
374
How do the locations of naïve, effector, and memory T cells differ?
* Naïve: Circulate through blood and lymph nodes. * Effector: Migrate to peripheral infected tissues. * Memory: Heterogeneous—some recirculate lymphoid organs, others reside in tissues.
375
What marks the lag phase in a primary T cell response?
Time from antigen encounter until clonal expansion (about days 0–7), before effector functions peak.
376
What two signals are required for naïve T cell activation?
1. Signal 1: TCR recognition of peptide-MHC. 2. Signal 2: Co-stimulation via CD28 on T cells binding CD80/CD86 on APCs.
377
Why is co-stimulation (Signal 2) necessary?
Ensures T cells activate only when APCs present antigen in an inflammatory/danger context, preventing responses to harmless or self-antigens.
378
Which molecules provide co-stimulation?
APCs express CD80/CD86 (B7-1/B7-2 in mice), which bind CD28 on T cells.
379
What intracellular kinase initiates TCR signal transduction?
Lck (lymphocyte-specific tyrosine kinase) phosphorylates ITAMs on the CD3 complex, leading to activation of transcription factors.
380
What is the role of IL-2 in T cell activation?
IL-2, produced after co-stimulation, drives autocrine clonal expansion (proliferation) of activated T cells.
381
What happens if a T cell receives Signal 1 without Signal 2?
It becomes anergic (non-responsive) or undergoes apoptosis, enforcing peripheral tolerance.
382
How do Tregs enforce peripheral tolerance?
Regulatory T cells express CTLA-4, which binds CD80/CD86 more strongly than CD28, blocking co-stimulation and inducing anergy in other T cells.
383
What clinical breakthrough targets CTLA-4 and PD-1?
Checkpoint inhibitors (anti-CTLA-4, anti-PD-1) unleash anti-tumour T cell responses; Nobel Prize 2018.
384
What is the “third signal” in T cell activation?
Cytokines from the same APC (and environment) that polarise activated T cells into functional subsets.
385
Name five CD4⁺ T helper subsets.
Th1, Th2, Th17, Treg, Tfh.
386
Which cytokine polarises Th1 differentiation?
IL-12 (and IFN-γ/IL-18).
387
Which transcription factor defines Th1 cells?
T-bet; Th1 cells produce IFN-γ, TNF-α, IL-2.
388
What functions do Th1 cells perform?
Activate macrophages and cytotoxic T cells for intracellular pathogen clearance (CMI).
389
Which cytokine polarises Th2 differentiation?
IL-4.
390
Which transcription factor defines Th2 cells?
GATA-3; Th2 cells produce IL-4, IL-5, IL-13.
391
What functions do Th2 cells perform?
Drive humoral responses against helminths and toxins; promote B cell class switching and eosinophil/mast cell activation.
392
Which cytokines polarise Th17 differentiation?
TGF-β, IL-1, IL-6.
393
What functions do Th17 cells perform?
Produce IL-17, IL-22; important for mucosal barrier defence and extracellular bacterial/fungal inflammation.
394
Which cytokines polarise Treg differentiation?
TGF-β, IL-2.
395
What functions do Tregs perform?
Produce IL-10, TGF-β to suppress/regulate immune responses and maintain self-tolerance.
396
Which cytokines polarise Tfh differentiation?
IL-6, IL-21.
397
What functions do Tfh cells perform?
Provide help to B cells in germinal centres, promoting affinity maturation and class switching (produce IL-4, IL-21).
398
How is cross-regulation between Th1 and Th2 achieved?
IFN-γ/T-bet from Th1 inhibits GATA-3/IL-4 of Th2; IL-4/GATA-3 from Th2 inhibits T-bet/IFN-γ of Th1.
399
What extra requirement do CD8⁺ T cells need for activation?
More co-stimulation from CD4⁺ Th cells—IL-2 production and CD40L–CD40 interactions to boost APC co-stimulation.
400
What is “cross-presentation”?
Exogenous antigens are presented on MHC I by APCs, enabling CD8⁺ T cell activation by pathogens not infecting the APC itself.
401
Summarise the three signals needed for full T cell activation.
1. TCR–peptide–MHC recognition 2. CD28–CD80/86 co-stimulation 3. APC-derived cytokines (polarisation)
402
What happens during the contraction/homeostasis phase?
After antigen clearance, co-stimulation wanes; ~95% of effector T cells die by apoptosis, leaving a small memory pool.
403
Describe the dynamics of primary vs. secondary T cell responses.
Primary response shows lag then peak around day 10; secondary response is faster/higher magnitude with minimal symptoms, lasting years.
404
What are the four steps of T cell extravasation into tissues?
1) Rolling via selectins, 2) Activation by chemokines, 3) Firm adhesion via integrins, 4) Diapedesis across endothelium.
405
Which selectins mediate T cell rolling?
E-selectin and P-selectin on inflamed endothelium binding their ligands on T cells.
406
How do chemokines guide T cells to infection sites?
Infected tissues secrete chemokines (e.g., CXCL10), creating a gradient that activates integrins and directs T cell migration.
407
How does an APC imprint tissue-homing properties on a T cell?
The APC’s cytokine and co-stimulatory context during activation induces specific adhesion molecules and chemokine receptors on the T cell.
408
What ensures a CD8⁺ T cell kills only infected targets?
Killing is MHC I–restricted and antigen-specific; the CTL must form a tight immunological synapse with the infected cell.
409
Name the two main cytotoxic mechanisms used by CTLs.
1) Perforin/granzyme–mediated apoptosis 2) Fas–FasL–induced caspase activation.
410
Why is apoptosis preferable to necrosis in CTL killing?
Apoptosis contains intracellular pathogens and prevents inflammation, avoiding collateral tissue damage.
411
What defines “unconventional” T cells?
They often use γδ or invariant αβ TCRs, recognise non-peptide antigens (lipids, metabolites), and reside in tissues (e.g., MAIT, iNKT).
412
What is the primary function of γδ T cells at epithelial barriers?
Rapid cytokine release (IFN γ, IL-17) and cytotoxicity against stressed or infected cells, maintaining barrier immunity.
413
How do iNKT cells recognise antigens?
Via CD1d-presented glycolipids, enabling early cytokine release (e.g., IL-4, IFN γ) in mucosal tissues.
414
What triggers the rapid response of MAIT cells?
MR1-presented microbial vitamin B₂ metabolites at mucosal surfaces, leading to IL-17/IFN γ production.
415
What proportion of effector T cells survive after antigen clearance?
Approximately 5% persist as long-lived memory T cells; ~95% die by apoptosis during contraction.
416
What mechanisms drive the contraction phase of the T cell response?
Loss of co-stimulation, upregulation of inhibitory receptors on T cells, and tissue wound-healing signals inducing apoptosis.
417
Why are memory T cells important?
They enable faster, larger secondary responses upon re-encounter with the same antigen, often preventing symptomatic reinfection.
418
Describe the three regions of the thymus and their roles in T-cell maturation.
* Cortex: Immature thymocytes undergo positive selection on cortical epithelial cells presenting self-MHC. * Corticomedullary junction: Intermediate thymocytes receive further signals. * Medulla: Negative selection by medullary epithelial cells and dendritic cells deletes high-affinity self-reactive T cells (central tolerance).
419
What defines “double-negative” and “double-positive” thymocytes?
* Double-negative (DN): CD4⁻CD8⁻ early precursors (DN1–4 stages) rearrange TCRβ. * Double-positive (DP): CD4⁺CD8⁺ cells express pre-TCR then rearrange TCRα before selection.
420
How does IL-2 receptor affinity change during T-cell activation?
* Naïve T cells: Express IL-2Rβγ (intermediate affinity). * Activated T cells: Upregulate CD25 (IL-2Rα) to form the high-affinity IL-2Rαβγ complex, driving robust proliferation.
421
Name the three major signalling pathways downstream of TCR engagement.
1. Calcineurin–NFAT (cytosolic calcium increase → NFAT activation) 2. Ras–MAPK (via LAT/Slack → AP-1 activation) 3. PKCθ–IKK (activates NF-κB).
422
What are the key adhesion molecules and integrins on T cells for tissue entry?
* Selectins: L-selectin (naïve homing to LN), E/P-selectin ligands (inflamed tissues) * Integrins: LFA-1 (CD11a/CD18) binds ICAM-1; VLA-4 (α4β1) binds VCAM-1 for firm adhesion.
423
How is L-selectin expression regulated upon T-cell activation?
Activated T cells shed L-selectin, reducing lymph node recirculation and promoting migration to peripheral tissues.
424
What are the defining features of central memory (T_CM) vs effector memory (T_EM) T cells?
* T_CM: CCR7⁺, CD62L⁺, home to lymphoid organs, proliferate vigorously upon re-challenge. * T_EM: CCR7⁻, CD62L⁻, patrol peripheral tissues, rapidly exert effector functions.
425
What are tissue-resident memory (T_RM) T cells?
Long-lived, non-circulating cells in peripheral tissues expressing CD69 and/or CD103, providing frontline defense at barrier sites.
426
How do antigen-presenting cells imprint gut-homing on T cells?
Gut dendritic cells produce retinoic acid, inducing α4β7 integrin and CCR9 on T cells for intestinal trafficking.
427
What metabolic shift occurs in T cells upon activation?
From oxidative phosphorylation in naïve cells to aerobic glycolysis (Warburg effect) in effectors, supporting biomass and rapid proliferation.
428
Which inhibitory receptors limit T-cell responses during contraction?
PD-1, CTLA-4, LAG-3 and TIM-3 upregulation dampens continued activation and promotes apoptosis of excess effectors.
429
How do memory T cells survive long term?
Homeostatic cytokines IL-7 and IL-15 support memory T-cell maintenance and low-level turnover in absence of antigen.
430
What is the role of CD40L on activated CD4⁺ T cells?
Binds CD40 on APCs to upregulate CD80/86 and cytokine production, enhancing CD8⁺ T-cell priming (“licensing”) and B-cell help.
431
Describe the sequential process of CTL degranulation.
1) Formation of immunological synapse, 2) Polarisation of MTOC, 3) Directed release of perforin/granzymes toward target cell.
432
What molecular change distinguishes naïve from activated LFA-1?
Inside-out signalling (via talin binding) shifts LFA-1 to a high-affinity conformation, enabling firm adhesion.
433
How does cytokine milieu shape Th17 vs iTreg differentiation?
* Th17: TGF-β + IL-6/IL-1 → RORγt → IL-17/IL-22. * iTreg: TGF-β + low IL-6 → FoxP3 → IL-10/TGF-β.
434
What transcriptional cross-inhibition occurs between T-helper subsets?
T-bet suppresses GATA-3; GATA-3 suppresses T-bet; RORγt and FoxP3 reciprocally inhibit each other for Th17/Treg balance.
435
What defines the immunological synapse in T-cell–APC interaction?
Central SMAC (TCR, CD28) and peripheral SMAC (LFA-1/ICAM) zones organising sustained signalling and adhesion.
436
How do CTLs deliver FasL to the target cell?
Upon synapse formation, CTLs upregulate FasL on their surface, engaging Fas on target to trigger caspase-8-mediated apoptosis.
437
Summarise how co-stimulatory strength influences T-cell fate.
* Strong, sustained CD28 signalling + high IL-2: Favors effector differentiation. * Weak/brief co-stimulation: Promotes memory precursor formation and long-lived memory.
438
What is the basic principle of the adaptive immune system?
Generate vast lymphocyte diversity to match pathogens, expand and refine matched clones, and maintain memory for future protection.
439
How do B-cell receptors (BCRs) differ from T-cell receptors (TCRs)?
BCRs are membrane-bound (or secreted as antibody), recognise intact antigen, and have class-determining Fc regions; TCRs bind peptide-MHC only on other cells.
440
List five effector functions of antibodies.
Neutralisation (toxins/viruses), opsonisation (phagocytosis), complement activation, agglutination, and antibody-dependent cell-mediated cytotoxicity (ADCC).
441
What enzymes mediate V(D)J gene rearrangement in B cells?
RAG1 and RAG2 (Recombination Activating Genes) catalyse combinatorial joining of V, D, and J segments in the bone marrow.
442
What segments compose the heavy-chain variable region?
V, D, and J gene segments (VDJ), plus the constant (C) region determining isotype.
443
What segments compose the light-chain variable region?
V and J segments (VJ), with either kappa or lambda light chains paired with heavy chain.
444
Explain combinatorial diversity mathematically.
Protein diversity = (#V)×(#D)×(#J); e.g., 6 V × 4 D × 3 J = 72 heavy-chain possibilities.
445
What is junctional diversity?
Addition (by TdT) or deletion of nucleotides at V–D–J junctions, boosting diversity beyond combinatorial potential.
446
Approximately how many different antibodies can humans generate?
On the order of 10 billion distinct specificities.
447
What two processes occur after B-cell activation to further diversify antibodies?
Somatic hypermutation and class-switch recombination, both requiring AID in the germinal centre.
448
What enzyme drives both somatic hypermutation and CSR?
Activation-Induced Cytidine Deaminase (AID).
449
How is class-switch recombination organised at the IgH locus?
Sequential Cμ → Cδ → Cγ3 → Cγ1 → Cα1 → Cγ2 → Cγ4 → Cε → Cα2 segments, recombined by AID to change isotype.
450
What are the functions of IgD?
Membrane receptor on naïve B cells; no secreted effector role.
451
What are the key features of IgM?
First isotype expressed; forms pentamers; potent complement activator; bulky, doesn’t cross placenta.
452
What distinguishes IgG subclasses?
Four subclasses (IgG1–4) differ in hinge length and Fc-mediated functions such as opsonisation and ADCC.
453
What are the roles of IgA?
Mucosal protection; secreted as dimer into secretions (milk, saliva); resists gut proteases.
454
How does IgE function?
Binds mast-cell FcεR in absence of antigen; crosslinking triggers degranulation; key in parasitic defense and allergy.
455
What defines TI-1 B-cell responses?
T-independent type I: polyclonal activation via pattern-recognition receptors (e.g., TLR ligands) without specific BCR engagement.
456
What defines TI-2 B-cell responses?
T-independent type II: repetitive antigen arrays (e.g., bacterial polysaccharides) crosslink many BCRs; minimal memory formation.
457
What defines T-dependent B-cell responses?
Protein antigens processed and presented on MHC II to CD4⁺ T-cells; require T-cell help for CSR, SHM, and memory formation.
458
What are the two signals in T-dependent B-cell activation?
“Signal 1”: BCR-mediated antigen uptake and presentation; “Signal 2”: CD40–CD40L interaction plus cytokines from T helper cells.
459
Which TNF-receptor family member is critical for germinal-centre formation?
CD40 (on B cells) interacting with CD40L (CD154) on T fh cells promotes proliferation, survival, and CSR.
460
Describe the germinal-centre dark and light zones.
Dark zone: centroblasts proliferate and hypermutate; light zone: centrocytes compete for antigen on FDCs and T fh help, undergoing selection.
461
How does affinity maturation occur?
Iterative cycles: SHM in dark zone → selection of higher-affinity BCRs in light zone → re-entry or differentiation into memory/plasma cells.
462
What fate choices arise from the germinal centre?
High-affinity B cells differentiate into long-lived plasma cells (antibody secretion) or memory B cells for rapid recall.
463
Where do short-lived extrafollicular plasma cells arise?
Outside germinal centres early in TD responses; secrete unmutated IgM before GC formation.
464
Why is vaccination important in humoral immunity?
It induces T-dependent GC responses, generating high-affinity, class-switched antibodies and durable memory B cells over time.
465
What is flow cytometry?
A technique that hydrodynamically focuses cells in suspension into a single‐cell stream, then measures light scattering and laser‐excited fluorescence to analyse cell properties at high throughput
466
What are the two non‐fluorescent parameters measured by flow cytometry?
* Forward scatter (FSC): proportional to cell size * Side scatter (SSC): proportional to internal complexity/granularity
467
How does hydrodynamic focusing work?
A high‐velocity sheath fluid surrounds the sample stream, aligning cells into a single file so each cell passes one at a time through the laser interrogation point
468
Why are fluorophore‐conjugated antibodies used in flow cytometry?
They bind specific cell markers; when excited by lasers, each fluorophore emits light at a characteristic wavelength, enabling multiplexed detection of multiple markers on the same cell
469
What determines multiplex capacity in a flow cytometer?
The number of lasers (excitation wavelengths) and detectors (emission filters) available; modern instruments can detect 12–28 colors, with some advanced systems detecting up to ~60
470
How are different fluorochromes distinguished?
Each fluorochrome has a unique excitation/emission spectrum; filters isolate emitted wavelengths so signals are attributed to the correct fluorochrome
471
What is an immunophenotyping “gate”?
A user‐defined region on a scatter or fluorescence plot that selects a cell subset of interest (e.g., lymphocytes by FSC/SSC) for further analysis
472
How do you identify T helper cells in a blood sample by flow cytometry?
1. Gate on lymphocytes (low FSC, low SSC) 2. Stain with anti-CD3 and anti-CD4 fluorophore-conjugated antibodies 3. Gate on CD3⁺CD4⁺ double-positive cells
473
Why must red blood cells be lysed before flow cytometry of whole blood?
RBCs lack nuclei and burst in lysis buffer (e.g., ammonium chloride), allowing removal of debris and haemoglobin while preserving more robust leukocytes for analysis
474
What is “autofluorescence” and how is it accounted for?
Natural fluorescence emitted by cells when excited; measured using an unstained control to set fluorescence gates above background
475
What is the purpose of an isotype control?
A fluorophore-conjugated antibody of the same species, isotype, and concentration but against an irrelevant antigen; measures non-specific binding via Fc receptors or sticky interactions
476
What tubes are needed for a basic immunophenotyping experiment?
* Unstained control: measures autofluorescence * Isotype control: measures non-specific binding * Test sample: contains specific fluorophore-tagged antibodies
477
What are common applications of flow cytometry?
* Immunophenotyping (e.g., CD4:CD8 ratio in HIV) * Live/dead assays * Cell cycle analysis * Cytokine production * Phagocytosis assays * Telomere length measurement
478
How is forward vs. side scatter used to distinguish leukocyte subsets?
* Lymphocytes: low FSC, low SSC * Monocytes: intermediate FSC, SSC * Granulocytes: high FSC, high SSC
479
Why are logarithmic scales used for fluorescence?
Because fluorescence signals vary over orders of magnitude; log scales spread out data to distinguish dim and bright populations
480
How can flow cytometry estimate protein expression levels, not just presence/absence?
Mean fluorescence intensity correlates with antigen density: brighter signal indicates higher protein expression per cell
481
What is electronic abort rate and why does it matter?
The percentage of events discarded when cells pass too close together; low abort rates (<1%) ensure accurate high‐throughput data (e.g., 3,000–10,000 events/sec)
482
How are population proportions calculated?
1. Determine % events in each quadrant of gated plot 2. Sum relevant quadrants for combined subsets (e.g., CD3⁺ = CD3⁺CD4⁺ + CD3⁺CD4⁻) 3. Calculate ratios or % of parent population
483
What factors can increase cell autofluorescence?
* Larger cell size (e.g., activated T cells) * Granularity (e.g., granulocytes) * Culture conditions (e.g., media components)
484
Why is panel design critical in flow cytometry?
To minimise spectral overlap, ensure each fluorochrome is excited by available lasers and detected with appropriate filters, and include necessary controls for compensation and gating
485
Why is a full blood count (FBC) important before flow cytometry?
It provides baseline counts and proportions of red and white blood cells; deviations can indicate disease and guide which subsets to analyse by flow cytometry.
486
How does flow cytometry characterise cells?
By hydrodynamically focusing cells in single‐file through a laser beam and measuring light scatter (size/granularity) and fluorophore‐emitted fluorescence for specific markers.
487
What are CD molecules and why are they used?
“Cluster of Differentiation” surface markers uniquely expressed on cell types (e.g., CD3 on T cells, CD4 on helper T cells); targeted by fluorophore‐conjugated antibodies for identification.
488
What is an isotype control and its purpose?
A fluorophore‐conjugated antibody of the same isotype but irrelevant specificity; measures non‐specific binding and sets accurate fluorescence gates.
489
Why must you incubate samples in the dark?
To prevent photobleaching of fluorophores, which reduces their fluorescence intensity and accuracy of detection.
490
What key information can you extract from your flow cytometry results?
Proportions and absolute counts of T‐cell subsets, cell size/granularity profiles, and assessment of staining specificity via isotype controls.
491
Why is immune tolerance necessary?
Random V(D)J gene rearrangements create diverse TCRs/BCRs, but can produce self-reactive lymphocytes that cause autoimmunity (“horror autotoxicus”) without tolerance mechanisms
492
What is central tolerance?
Deletion or editing of self-reactive T cells in the thymus and B cells in the bone marrow before they enter circulation
493
Where and how does positive selection of T cells occur?
In the thymic cortex, thymocytes that bind self-MHC weakly receive survival signals; non-binders die by apoptosis
494
Where and how does negative selection of T cells occur?
In the thymic medulla, medullary epithelial cells and DCs present self-antigens; thymocytes with high-affinity self-MHC binding undergo apoptosis
495
How do immature B cells achieve central tolerance?
In the bone marrow, self-reactive B cells may: 1. Undergo receptor editing (light-chain recombination) 2. Die by apoptosis if editing fails 3. Become anergic if binding is weak
496
What are the limitations of central tolerance?
Not all self-antigens are expressed in primary lymphoid organs; deleting all self-reactive cells would overly narrow the repertoire; environmental antigens aren’t encountered centrally
497
What is peripheral tolerance?
Mechanisms acting on mature T and B cells in the periphery to control any self-reactive cells that escaped central tolerance
498
Describe “ignorance” in T-cell peripheral tolerance.
Some self-antigens are hidden (sequestered) or expressed at low levels and never reach activation threshold, so T cells remain unresponsive
499
What causes T-cell anergy?
TCR engagement without co-stimulation (CD28–CD80/86) or active inhibition by CTLA-4, leading to unresponsiveness on restimulation
500
How does CTLA-4 enforce peripheral tolerance?
Upregulated after T-cell activation; binds CD80/86 with higher affinity than CD28, sending inhibitory signals and suppressing APCs
501
What is Activation-Induced Cell Death (AICD)?
Repeated TCR stimulation induces Fas/FasL and pro-apoptotic proteins (Bax, Bad, Bim), triggering apoptosis of self-reactive T cells in the periphery
502
What are the two main actions of regulatory T cells (Tregs)?
* Contact-dependent: High CTLA-4 expression binds APCs, can directly kill via perforin/granzyme * Soluble factors: Secrete IL-10 and TGF-β; “mop up” IL-2 via low-affinity IL-2R
503
What distinguishes natural vs. inducible Tregs?
* Natural Tregs: Develop in the thymus, express FoxP3 intrinsically * Inducible Tregs: Arise in periphery from naïve CD4⁺ T cells upon antigen + tolerogenic signals
504
How is B-cell peripheral anergy induced?
Chronic low-level antigen and BCR cross-linking without adequate co-stimulation (e.g., CD40L, TLR signals) renders B cells unresponsive
505
How does acute antigen exposure lead to B-cell deletion?
High-level antigen cross-linking without T-cell help triggers B-cell apoptosis via intrinsic death pathways
506
What is the “exclusion” mechanism for B-cell tolerance?
Partially activated B cells fail to express homing chemokine receptors, so cannot enter follicles or receive survival signals to mature
507
What additional factor regulates peripheral B-cell tolerance?
FcγRIIB engagement by immune complexes delivers inhibitory signals; limited BAFF (survival factor) can also cause B-cell death
508
How does a breakdown of tolerance lead to autoimmune disease?
Failure in central or peripheral tolerance allows self-reactive lymphocytes to persist and attack tissues, mediated by hypersensitivity II–IV mechanisms
509
What genetic and environmental factors contribute to autoimmunity?
Susceptibility genes (e.g., HLA-DQ/DR alleles) plus triggers (infections, hormones, gut dysbiosis) combine to breach tolerance
510
Give an example of an immunologically privileged site.
The anterior chamber of the eye or areas behind the blood–brain barrier, where antigens remain sequestered from immune surveillance
511
Why might some self-reactive cells be beneficial?
They contribute to tumor surveillance and help regulate normal inflammatory responses, balancing immunity with tolerance
512
What is “hypersensitivity”?
An excessive or misdirected normal immune response that causes tissue damage when regulatory mechanisms fail or responses occur at the wrong time
513
How are hypersensitivity reactions classified?
By the Gell & Coombs system into four types: * Type I: IgE-mediated (immediate) * Type II: IgG/IgM-mediated (antibody against cell/tissue antigens) * Type III: Immune complex-mediated * Type IV: T cell-mediated (delayed)
514
Which hypersensitivity types are antibody-mediated, and which is cell-mediated?
* Antibody-mediated: Types I, II, III * Cell-mediated (delayed): Type IV
515
Outline the sensitisation and elicitation phases of Type I hypersensitivity.
1. Sensitisation: First exposure → Th2-driven B cells class-switch to IgE → IgE binds FcεRI on mast cells. 2. Elicitation: Re-exposure → Allergen cross-links mast-cell IgE → Immediate degranulation and mediator release
516
What key mediators are released by mast cells in Type I reactions?
* Pre-formed: Histamine, proteases * Newly synthesized: Prostaglandins, leukotrienes, cytokines (e.g., TNF)
517
Which cytokines from Th2 cells promote IgE production?
IL-4 (class switching to IgE) and IL-5 (eosinophil activation)
518
Give three clinical examples of Type I hypersensitivity.
Allergic rhinitis (hay fever), Asthma, Systemic anaphylaxis
519
Describe the mechanism of Type II hypersensitivity.
IgG or IgM auto- or allo-antibodies bind cell-surface or matrix antigens → Fc receptor–mediated phagocytosis/ADCC and/or complement fixation → tissue injury
520
What is the difference between Type II and 'Type V' hypersensitivity?
Type II: Antibody-mediated cell/tissue destruction Type V: Antibody-mediated functional modulation of receptors (e.g., Graves’ disease)
521
Provide two examples of Type II cytotoxic reactions.
Autoimmune haemolytic anaemia (anti-RBC antibodies → phagocytosis/MAC), Goodpasture’s syndrome (anti-GBM IgG → glomerulonephritis)
522
Give two examples of Type II functional (Type V) reactions.
Graves’ disease (anti-TSH-receptor → hyperthyroidism), Myasthenia gravis (anti-AChR → muscle weakness)
523
How are immune complexes handled, and why do they cause Type III reactions?
Normally phagocytosed by macrophages; excess small complexes deposit in vessel walls → complement activation + neutrophil recruitment → inflammation and tissue damage
524
Contrast the tissue sites favored by small vs. large immune complexes.
Small complexes: Deposit in blood vessels → vasculitis Large complexes: Cleared in liver/spleen; may accumulate in kidneys if complement deficient
525
Name two classic Type III hypersensitivity diseases.
Serum sickness (e.g., anti-snake venom), Systemic lupus erythematosus (anti-DNA immune complexes → glomerulonephritis)
526
What characterises Type IV (delayed-type) hypersensitivity?
Sensitised T cells (mainly Th1) release IFN-γ on re-exposure (1–2 weeks) → macrophage activation and/or CTL-mediated cytotoxicity → tissue damage
527
What is a hapten, and how does it cause contact dermatitis?
A small molecule that binds host proteins → presented on MHC I/II → CD8⁺ CTLs kill cells and Th1 cells activate macrophages → rash (e.g., poison ivy)
528
Describe granulomatous (Type IV) hypersensitivity.
Persistent antigen (e.g., Mycobacterium tuberculosis) → Th1-driven IFN-γ activates macrophages → fused “giant” cells form granuloma to contain infection
529
List three diseases featuring granulomatous hypersensitivity.
Tuberculosis, sarcoidosis, Crohn’s disease
530
Why is the DTH reaction 'delayed'?
T cell priming and recruitment of macrophages take 24–72 hours to develop inflammation upon antigen re-exposure
531
Summarise the four hypersensitivity types with their principal effector and time course.
I: IgE & mast cells, immediate (<1 hr) II: IgG/IgM & complement/ADCC, hours III: Immune complexes & neutrophils, hours IV: T cells & macrophages/CTLs, delayed (1–2 days)
532
What characterises the late‐phase reaction in Type I hypersensitivity?
Occurs 4–12 hours after initial mast-cell degranulation; driven by newly synthesized mediators (e.g. leukotrienes, cytokines) causing sustained inflammation (e.g. bronchospasm, edema).
533
Name three pharmacologic blocks of Type I hypersensitivity.
H₁‐antagonists (e.g. diphenhydramine) block histamine-mediated itch/vasodilation Leukotriene receptor antagonists (e.g. montelukast) reduce bronchoconstriction Epinephrine reverses laryngeal edema and hypotension in anaphylaxis
534
Which histamine receptor mediates bronchoconstriction and vascular permeability?
H₁ receptors on smooth muscle and endothelium.
535
What is the Arthus reaction?
A localized Type III vasculitis seen after intradermal injection of antigen in pre-immunized individuals; immune complexes form in vessel walls causing edema, hemorrhage.
536
How does serum‐sickness differ from the Arthus reaction?
Serum sickness is systemic—large quantities of antigen (e.g. antitoxin) generate immune complexes that deposit in vessels, joints, and kidneys days after injection.
537
Why do small immune complexes deposit more easily than large ones?
Small complexes are less efficiently cleared by spleen/liver macrophages and can slip through endothelium into tissues.
538
Which complement pathway is most critical in Type III damage?
The classical pathway, initiated by C1 binding IgG/IgM in complexes, generates C5a to recruit neutrophils.
539
What are the four subtypes of Type IV hypersensitivity?
IVa: Th1-macrophage (e.g. PPD test) IVb: Th2-eosinophil (e.g. chronic asthma) IVc: CTL-mediated (e.g. Stevens–Johnson syndrome) IVd: T-cell–neutrophil (e.g. acute generalized exanthematous pustulosis)
540
How is a Mantoux (PPD) test read?
Intradermal PPD injection; induration measured at 48–72 h. ≥5–15 mm of firm swelling indicates prior sensitization.
541
What is contact dermatitis and its timeline?
A Type IVa reaction to haptens (e.g. nickel) on skin—erythema and vesicles develop 24–48 h after contact.
542
Which cytokine is the principal driver of granuloma formation in IV hypersensitivity?
IFN-γ from Th1 cells, activating macrophages to wall off persistent antigens.
543
How does penicillin cause a Type II reaction?
It acts as a hapten, binding RBCs—and anti‐penicillin antibodies mediate complement lysis (hemolytic anemia).
544
What laboratory test detects immune complexes in serum‐sickness?
C1q binding or CH50 assay—low complement levels indicate ongoing consumption by complexes.
545
Which cells execute tissue damage in Type IVc reactions?
CD8⁺ cytotoxic T lymphocytes via perforin/granzyme (e.g. in Stevens–Johnson syndrome).
546
Summarise immediate vs delayed cutaneous hypersensitivity testing.
Immediate (Type I): Skin‐prick test read in 15–20 min for wheal‐and‐flare (IgE) Delayed (Type IV): Patch or intradermal test read at 48–72 h for induration (T cells).
547
Why are mucosal surfaces a special immunological compartment?
They’re the major entry points for pathogens (gut, lung, genitourinary tracts), require discrimination between harmful microbes and harmless antigens, and host ~80% of all immune cells.
548
What does MALT stand for and what are its subdivisions?
Mucosa-Associated Lymphoid Tissue, including GALT (gut), BALT (bronchial), and NALT (nasopharynx).
549
What dual functions does the mucosal immune system perform?
1) Defence against invasive pathogens, 2) Prevention of responses to commensal flora and food antigens (mucosal tolerance).
550
How does the site of antigen entry dictate the lymphoid response?
Blood → spleen; Skin → draining lymph nodes; Mucosa → MALT (e.g., Peyer’s patches for gut antigens).
551
Name three organized lymphoid structures in the GI tract.
Peyer’s patches, appendix, and scattered (pseudo-follicles) intraepithelial lymphocytes.
552
What are the three layers of protection at mucosal surfaces?
1) Physical barrier (mucus, epithelium), 2) Innate immunity (DCs, macrophages, ILCs), 3) Adaptive immunity (sIgA, Th17, Tregs).
553
What components make up the mucus barrier in the gut?
Glycoproteins (e.g., MUC2) from goblet cells, antimicrobial peptides (e.g., defensins) from Paneth cells, plus secretory IgA.
554
How do epithelial tight junctions contribute to mucosal defence?
They maintain selective permeability, preventing most microbes and large molecules from crossing into tissue.
555
What is the role of innate lymphoid cells (ILCs) in steady-state mucosa?
Attracted by epithelial MIP3α, they help maintain barrier integrity and secrete cytokines upon activation.
556
How do mucosal DCs differ from systemic DCs?
They extend dendrites between enterocytes to sample lumenal antigens, produce IL-10/TGF-β (tolerogenic), and promote IgA class-switching and Treg differentiation.
557
What triggers mucosal DCs to become inflammatory?
Sustained TLR signals from invasive pathogens; they then secrete IL-1, IL-6, TNFα and recruit ILCs and Th17 cells.
558
What are the two functional zones of a Peyer’s patch?
Inductive site (antigen uptake/activation) and effector site (armed lymphocytes migrate to lamina propria).
559
What specialized epithelial cell transports antigen into Peyer’s patches?
M (microfold) cells, which transcytose antigen from the lumen to sub-epithelial APCs.
560
Outline the sequence of adaptive activation in a Peyer’s patch under homeostatic conditions.
M cells → DCs capture antigen → conditioned by epithelial TGF-β → DCs induce Th2/Tregs → B cells class-switch to IgA → plasma cells migrate to lamina propria.
561
What signals drive B-cell class-switch to IgA in GALT?
TGF-β, IL-10 and CD40L from Tfh/Tregs in germinal centres.
562
How is polymeric IgA transported across epithelium?
Dimeric IgA binds pIgR on basolateral surface, is transcytosed, and pIgR is cleaved to release secretory component–bound sIgA.
563
What functions does secretory IgA perform?
Neutralises pathogens/toxins, prevents microbial adherence, aids clearance, without fixing complement or inducing inflammation.
564
Why is IgA deficiency often asymptomatic?
Compensation by other isotypes (e.g., IgM) and intact physical/innate barriers.
565
Describe the mucosal response to invasive bacteria.
Epithelial TLRs → IL-1, IL-6, TNFα → DC/ILC1 activation → Th17 differentiation (with TGF-β, IL-6, IL-1) → IL-17A/IL-22 secretion → neutrophil recruitment and AMP production.
566
What are the key effector cytokines of Th17 cells?
IL-17A (neutrophil recruitment/activation) and IL-22 (antimicrobial peptides, increased permeability).
567
How does the mucosal response differ for parasitic infections?
Dominant Th2/ILC2 response (IL-4, IL-5, IL-13) leading to eosinophil recruitment and barrier repair.
568
What maintains mucosal tolerance to food antigens?
High Treg/IL-10/TGF-β environment, tolerogenic DC conditioning, and sIgA–mediated exclusion.
569
What is the role of epithelial-derived TGF-β in mucosal immunity?
Conditions DCs to a tolerogenic phenotype, promotes IgA class-switching and Treg differentiation.
570
How do mucosal macrophages sample antigen without disrupting barrier integrity?
They extend processes between tight-junctioned epithelial cells into the lumen to capture pathogens.
571
What three cell types dominate the lamina propria effector site?
Plasma cells (sIgA), CD4⁺/CD8⁺ T cells, and innate lymphoid cells.
572
Why is Crohn’s disease considered a breakdown of mucosal homeostasis?
Genetic susceptibility + abnormal microbe interactions → excessive Th1/Th17 inflammation, granuloma formation, transmural lesions.
573
Contrast Crohn’s disease vs ulcerative colitis pathology.
Crohn’s: Any GI region, transmural inflammation, granulomas UC: Colon only, mucosal layer, crypt abscesses, cancer risk.
574
List environmental factors implicated in IBD.
Western diet, altered microbiota, vitamin D deficiency, smoking, medications.
575
How do monogenic and polygenic factors contribute to IBD?
HLA haplotypes and polymorphisms in epithelial/immune genes increase susceptibility; family/twin studies show heritability.
576
What chemokine attracts all ILC subsets to the epithelium?
Epithelial cells secrete MIP3α, which recruits ILC1, ILC2, and ILC3 to maintain barrier integrity.
577
How do monogenic and polygenic factors contribute to IBD?
HLA haplotypes and polymorphisms in epithelial/immune genes increase susceptibility; family/twin studies show heritability
578
What chemokine attracts all ILC subsets to the epithelium?
Epithelial cells secrete MIP3α, which recruits ILC1, ILC2, and ILC3 to maintain barrier integrity
579
What are the main functions of ILC1, ILC2, and ILC3?
ILC1: Produce IFN-γ, mirror Th1, defend against intracellular pathogens ILC2: Produce IL-5/IL-13, mirror Th2, drive anti-helminthic responses and tissue repair ILC3: Produce IL-17/IL-22, mirror Th17, promote neutrophil recruitment and antimicrobial peptide production
580
How do M cells deliver antigen to Peyer’s patches?
M cells express PRRs (e.g., TLRs) on their apical surface, take up lumenal antigen by transcytosis, and hand it to sub-epithelial DCs/macrophages
581
How do epithelial TLR signals shape mucosal DCs?
TLR-activated enterocytes release TGF-β and growth factors that “condition” DCs to a tolerogenic phenotype, promoting Treg induction and IgA switching
582
Outline the steps of pIgR-mediated IgA transport.
1. Dimeric IgA (with J chain) binds pIgR on basolateral epithelium 2. The complex is transcytosed 3. Proteolytic cleavage releases sIgA with its secretory component into the lumen
583
What proportion of total body Ig is IgA, and how much is secreted daily?
Over 75% of body immunoglobulin is IgA, with around 3 g secreted into mucosae each day
584
Why is sIgA considered “non-inflammatory”?
sIgA does not fix complement, activate granulocyte degranulation, or induce full DC maturation—its role is homeostatic exclusion and neutralisation without inflammation
585
What genetic factors predispose to IBD?
HLA-DQ/DR haplotypes Polymorphisms in epithelial and immune genes Monozygotic twin concordance ~50% for Crohn’s, ~20% for UC
586
Which environmental factors contribute to IBD risk?
Western diet, altered microbiota, vitamin D deficiency, certain medications, and smoking all increase susceptibility
587
Contrast Crohn’s disease vs. ulcerative colitis pathology.
Crohn’s: Transmural lesions, granulomas, anywhere in GI (often ileum), Th1/Th17-driven, anti-bacterial IgG UC: Mucosal-only inflammation, crypt abscesses, colon-restricted, mixed Th1/Th2, increased carcinoma risk
588
How many V, D, and J gene segments compose the human Ig heavy-chain locus?
Approximately 61 V, 27 D, and 6 J segments, recombined to form heavy-chain variable regions
589
What are the numbers of V and J segments in human kappa and lambda light-chain loci?
Kappa: ~48 V and 5 J segments; Lambda: ~40 V and 7 J segments
590
Describe the framework (FW) and complementarity-determining region (CDR) organization of the Ig variable domain.
FW1–CDR1–FW2–CDR2–FW3–CDR3: CDR3 (formed at V–(D)–J junction) is the most hypervariable for antigen binding
591
What two processes add junctional diversity and roughly how?
* Exonucleolytic trimming removes random nucleotides at V/D/J ends * TdT-mediated N-region addition inserts random nts at junctions
592
Roughly how many distinct BCR specificities can combinatorial + junctional diversity generate?
Estimates ~10 billion possible unique receptors
593
Which enzyme and microenvironment drive both somatic hypermutation and class switching?
Activation-induced cytidine deaminase (AID) in germinal-centre B cells
594
Contrast extrafollicular vs germinal-centre B-cell responses.
* Extrafollicular: Rapid, short-lived IgM plasma blasts, no SHM or CSR * Germinal-centre: Slower, high-affinity, class-switched memory/plasma cells via SHM & CSR
595
In the germinal centre, what defines the “dark” vs “light” zones?
* Dark zone: Centroblasts proliferate and hypermutate * Light zone: Centrocytes compete for antigen on FDCs and Tfh help, undergoing selection
596
How does sequential class-switch recombination enforce directionality?
AID excises intervening constant genes; once downstream isotypes (e.g. IgA2) are reached, upstream ones cannot be re-acquired
597
What distinguishes TI-1 from TI-2 B-cell activation?
* TI-1: Polyclonal activation via TLRs (e.g. CpG), independent of BCR specificity * TI-2: Repetitive non-protein antigens (e.g. bacterial polysaccharides) crosslink many BCRs; minimal memory
598
Outline the two-signal requirement for T-dependent B-cell activation.
1. Signal 1: BCR binds and processes protein antigen → pMHC II presentation 2. Signal 2: CD40L on T fh engages CD40 on B cell + cytokines
599
What key role do follicular dendritic cells (FDCs) play in the light zone?
Display native antigen–antibody complexes for centrocyte BCR testing, guiding selection of high-affinity clones
600
How do memory B-cell and long-lived plasma-cell outputs differ post-GC?
* Memory B cells: Recirculate, rapidly re-enter GCs on rechallenge * Plasma cells: Migrate to bone marrow niches to continuously secrete high-affinity Ab
601
In the Moderna SARS-CoV-2 vaccine study, when did circulating plasmablasts and memory B cells peak?
* Plasmablasts: Peak ~Day 7 after 1st dose (extrafollicular) * Memory B cells: Peak ~Day 7 after 2nd dose (GC-derived)
602
Why is the germinal-centre reaction described as “Darwinian selection”?
B-cell clones hypermutate randomly; only those with improved affinity capture antigen/Tfh help and survive to become memory/plasma cells