Immunology Flashcards

1
Q

Give examples of mechanical innate imune defences

A
  • epithelial cells joined by tight junctions
  • longitudinal flow of air/fluid
  • mucus movement by cilia (lungs)
  • tears & nasal cilia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Give examples of chamical innate immune defences

A
  • fatty acids
  • B-defensins, lamellar bodies, cathelicidin
  • low pH/enzymes (pepsin)
  • a-defensins, lecticidins
  • pulmonary surfactant
  • tear/saliva enzymes (lysozymes)
  • histatins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

List some innate immune cells

A
  • NK cells
  • dendritic
  • monocytes

Granulocytes:
- neutrophils
- eosinophils (immune cell recruitment)
- basophils
- mast cells

Alle except NK derived from myeloid progenitor, Nk from lymphoid progenitor

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

Macrophage function

A

Phagocytosis & activation of bactericidal mechanisms.
Antigen presentation.

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

Monocyte function

A

Found in blood - can enter tissue + differentiate into macrophage

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

What are the different phaocyte receptors?

A
  • Mannose (C-type lectin R binds carbs)
  • Dectin-1 (B-glucan rec), anti-fungal C-type lectin R
  • scavenger, binds low density lipoproteins (sialic acid) on bacteria + yeast
  • lipid, directs immune function & metabolism
  • Complement (CR3, CR4), recognises LPS/pipophosphoglycans on bacteria/yeast

Also toll-like receptors

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

What are toll-like receptors?

A

Pattern recognition receptor family - recognises very conserved structures.

On pathogens: pathogen associated molecular patterns (PAMPs)

On damaged cells : danger associated molecular patterns (DAMPs)

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

Give examples of TLRs for bacteria and viruses

A

Bacteria - TLR9 recognises unmethylated CpG motifs -> MyD88 adaptor protein activation (cytokine production)

Viruses - TLR3 recognises dsRNA, signals TRIF pathway (TBK1) -> induces IRF3 & NF-kB, IFN-B production
(complex isgnalling cascade)

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

Describe 3 other pattern recognition receptor families

A

RLR - retinoic acid inducible gene (RIG) like receptors e.g. RIG-1 for viral recognition

CLR - C-type lectin receptors, major role in yeast recognition (some bacteria + parasitic worms)

NLR - nucleotide binding domain (NOD) like receptors e.g. T1 + NLRP3, cytosolic receptors

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

How do damage receptors on phagocytes work?

A

Recognise damage caused by pathogens - allows co-existence w/ beneficial microbes.

  • RAGE binds calprotectin (inflam marker)
  • NLRP1, NLRP3 & NLRC4
  • P2X7R binds ATP -> ASC/NLRP3 inflammasome assembly, also releases IL-1B & IL-18

RAGE: Receptor for Advanced Glycation End-products

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

List the names of cytokine and their general function

A

Soluble, hormone-like factors in the immune system

Interleukins (IL1-41) - diverse
Interferons (IFN) - anti-viral
Colony stimulating factors - haematopoesis
Tumour necrosis factor (TNF) - inflammation
Chemokines - chemotaxis

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

What is the inflammasome?

A

Complex of signalling molecules which:
- initiate type of programmed cell death
- enable secretion of active cytokines (IL-1B, IL-18)

Initiated by NLRs (e.g. NLRP1/3, NLRC4)

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

Describe key features of inflammation (local)

A

Can be both local & systemic.

Local:
- Vasodilation, enhanced vascular permeability -> mediated by TNF-a, IL-1, IL-6, histamine (released by mast cells)
- Oedema from accumulated plasma fluid + proteins
- Neutrophils + monocytes recruited via chemotaxis through permeable vascular system (immune amplification)

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

Mast cell function

A

Has granules - rapidly deployed and degranulated when activated.
Can transcirbe new products.
Immediate - histamine, herpin + enzymes (tryptase & chymase)
Delayed - prostaglandins, leukotrienes & cytokines

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

Systemic inflamatory response

A

If local cytokine response high enough - endocrine signalling.

IL-6, IL-1B & TNF-a stimulate hepatocytes to make acute phase proteins.

Il-1 in brain can promote fever, anorexia & somnolence

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

What are opsonins?

A

Acute phase proteins - ‘eat me’ signals for macrophages (phagocytosis)
- can also trigger complement

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

Key opsonins and their binding target

A

C-reactive protein (CRP) binds phosphoryl choline on microbes + damaged cells.

Mannose-binding lectin (MBL) binds mannose-fucosyl residues.

C3b cleaved from comp C3 binds -OH or -NH2 on anything (inactivated by host cells)

Surfactant proteins (SP-A & SP-D) bind various inc. LPS on microbes inc. viruses.

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

What is complement the 3 cascades it can induce?

A

Collection of soluble proteins that circulate in an active form - when triggered initiate activation cascde:
1. classical (CRP trigger)
2. Alternative (C3b trigger)
3. Lectin (MBL trigger)

Targets -> lysis, chemotaxis, mast cell activation, immune complex clearance

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

Interferon function

A

Inflam cytokine.
Inhibits viral replication, binds IFN-Rs -> renders resistant to infection - also activates macrophages & NKCs

Also secreted by infected ceel so acts in autocrine & paracine manner.

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

Natural kiler cells

A

Release lytic granules tha destroy virus infected cells -> critical for early IFN producction

Lymphoid like

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

How many families of innate lymphoid cells are there?

A

3 - ILC1-3

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

Primary lyphoid organs

A

Lymphocytes generated & mature
- bone marrow
- thymus

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

Secondary lymphoid organs

A

Lymphocytes encounter antigens & intitiate immune response.
- spleen
- lymph nodes
- tonsils
- Peyer’s patches

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

B cell receptor structure

A

2x Light chain + heavy chain -> variable & constant regions (membrane immunoglobin)

Also has adjacent IgB/Iga heterodimer (crucial for localisation to cell surface

Antigen binding triggers phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) on CD79a/CD79b subunits

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Signalling function of BCRs
Syk and Lyn kinases recruited + actvivated to form signalosome -> can activate various downstream signalling pathways (RAS/RAF & Nf-KB/NFAT) - Signalling determines fate of antigen-encountered B cells - Modulates gene expression, adhesion or survival - Differentiate into plasma B cell ## Footnote Syk mediates IP3 generation, whereas Lyn regulates Ca2+ mobilization through a process independent of IP3 generation.
26
Stages of antibody production by B cells
Produced on B cell surface or secreted (plasma cells) - antigen selectively activates virgin lymphocyte - clonal proliferation - maturation into effector plasma cells & memory cells
27
Describe the basic structure of an antibody
Heterodimers: 2 identical light (25 kDa) + 2 identical heavy (50 kDa) chains joined by disuphide bonds. 2 Fab regions (fragment antigen binding) Fc - fragment crystallisable -> effector domain Papain can cleave disulphide bonds linking heavy chains (Fab can bind antigen but Fc does not - but can be crsytallised) ## Footnote Immunglobinds so have Ig domain
28
What are the binding domains of antibodies?
N-terminal, have variable (H) and variable (L) at each binding site -> Fv Hinge region -> flexibility for antigen binding Hypervariable regions form complementary dtermining region (CDR) - mst be comp to antigenic epitope
29
What are antigens?
Immune response inducing molecule recognised by Abs - commonly proteins but can be polysaccahrides, lipids, DNA etc. Epitopes - fragments on Ag recognised by Ab e.g. SARS-Cov2 epitopes are spike proteins - an antigen can have multiple different epitopes
30
Linear vs conformational epitopes
Conformational: - Structural, formed by protein folding but may not be a continuous sequence of a. acids. - Most commonly recognised by Abs Linear: - Continuous sequence of a. acids - Less common - used in Western Blotting after protein denaturation
31
Can the same Ab recognise different antigens?
Yes - may share seuqence/structural homology, common epitopes. Myriad of antibodies produced during infection to recognise many different epitopes
32
Affinity
Strength of binding between an epitope & an antibody antigen binding site. - Interactions are weak unless 2 binding mols are close together ## Footnote Binding forces are non-convalent - varied affinities
33
How is cross-reactivity of an antibody generated?
Same Ab can bind different epitopes - affinity will be less than for original.
34
Avidity
Strength of an Ab-Ag complex. - If an antibody is using both binding sites to bind 2 epitopes on same antigen then avidity is increased Repeating epitopes can increase potential avidity
35
Antibody vs innate immune responses
Antibody: - acquired after pathogen exposure - pathogen specific - memory -> 2nd response bigger + faster Innate: - immediate, pre-existing - Non-specific for any pathogen - No memory: 2nd response same as 1st
36
List 4 antibody functions
- neutralisation - opsonization - complement activation - Ab dependent cell mediated cytotoxicity (ADCC)
37
What is neutralisation? ## Footnote Ab function
- Antibodies prevent pathogen/protein binding to their target - Important in protection against virus, prevents entry into cell + replication e.g. vaccines ## Footnote All 4 IgGs & IgA
38
What is opsonization? ## Footnote Ab function
Pathogen is tagged by antibodies for phagocytosis by macrophage/neutrophils. - Fc domain binds Fc receptor on phagocyte -> trigger ## Footnote IgG1, IgG3
39
What is complement activation? ## Footnote Ab function
Antibodies attached to surface of pathogen activate 1st protein in complement system. Can lead to: 1) Deposit complement proteins on surface of bacterium -> leads to pore formation + lysis of bacterium directly 2) Complement receptors on phagocytes recognise these comp proteins + induce phagocytosis ## Footnote IgM, IgG1, IgG3
40
What is antibody dependent cell mediated cytotoxicity (ADCC)? ## Footnote Ab function
Viral infected host cells can express viral surface proteins. - recognised by specific Abs - NKCs/macrophages w/ Fc receptors bind Ab (cross-link), killing target cell by perforin + granzymes release ## Footnote Mediates extracellular killing via apoptosis - IgG1 & IgG2
41
What process does blood-type determination rely on?
Agglutination - Ab ability to immunoprecipitate a coluble or non-soluble antigen A & B transferases encoded by ABO gene. Agglutination + hemolysis of RBC caused by variable antibody binding. ## Footnote AB is universal recipient, O is universal donor
42
What are different antibodies designed for?
Type depends on C region of heavy chains - class specific. Designed for - bindinf Fc receptors, complement actvation, secretion regulation.
43
What are some different Ab types?
IgG: most common, 4 classes, IgG1 most common IgM: earliest Ab made after antigen contact, pentamer via J chain & disulphide bonds, up to 10 binding sites IgD: rare, function unclear IgA1: serum & major Ig in mucosa, 2 subclasses, different structure in secretions: 2 Ig mols joined IgE: lowest in serum, activates mast cells, important for allergies + worm infection
44
Which antibodies form polymers?
IgM - pentameric via J chain & disulphde bonds (up to 10 binding sites) IgA - dimeric, 2 Ig mols joined via J chain
45
Key characteristics of IgA
Useful for transport from lamina propria across epithelium into gut. - bind to receptor on basolaterla face of ep cell - endocytose - transcytosis to apical face of ep cell - IgA dimer + secretory component released at apical face of ep cell ## Footnote Dimers (IgA) predominant in external secretion, at mem surfaces
46
What is mucosal associated lymphoid tissues (MALT)?
Another secondary lymphoid tissue - site of B cell activity (esp in gut)
47
What are the two main types of T-lymphocytes?
CD8 - Tc/CTL CD4 - Th ## Footnote Both from thymus - declining function w/ age (it involutes)
48
Structure of T cell receptor
a & B chain - variable + constant regions. Also has hinge (linked by S-S), TM & cytoplasmic tail domains (ITAMs) Has CDR from vriable region that binds antigen. Associated with CD3 receptors - mediates intracellular signalling
49
How do TCRs recognise antigens
MUST be presented via APC, indirect binding to processed antigen presented by MHC mol
50
What is a major histocompatability complex (MHC)?
Antigen presenting molecule - coded by genes arrayed in long continuous DNA sgement. HLA (human leukocyte antigen) in humans, mouse H-2, rats RT1. They binds antigenic peptides & present them on surface to T cells.
51
MHC-I structure
- long a-chain (3 domains) bound to membrane + non-covalently linked to 2B chain - Only 1 TM domain (a-chain) - found on most nucleated cells It binds 8-10 a.acid peptides, direct CD8+ response
52
MHC-II structure
- shorter a-chain non-covalently linked to equal B-chain - 2 TM domains from both chains - restricted expression (macrophages, B cells, dendriitc cells) Binds 12-20+ a.acid peptides, direct CD4+ response
53
# Think genetics What are two main features of MHC molecules?
Polygenic - several different class I & II genes (III not for antigen presentation) contribute to phenotype Polymorphic peptide binding region - multiple variants of each gene within population -> hundreds of forms means different phenotypes
54
# ``` ``` How are MHC-I and MHC-II polygenic?
MHC-I: - a-chain encoded by A, B & C loci in humans - B2-microglobulin encoded by gene on different chrom (12 instead of HLA on Chr6) MHC-II: - a & b-chains encoded by DP, DQ & DR regions of HLA in humans
55
Co-dominant expression of MHC genes
Inherent maternal + paternal gene products expressed equally in a cell SO most individuals are heterozygous -> advantageous for processing wider antigenic range. ## Footnote Processed Ag peptides are small fragements (7-18 a.acids)
56
Processing of endogenous antigens to MHC-I
Proteosome degrades antigen + peptides transported by TAP to rER. p88 (calnexin) dissociates from MHC-I in rER when peptide bound | TAP - transporter associated with antigen processing ## Footnote Class-I -> presentation of intracellular antigens (viral)
57
Formation of MHC-I in ER
- partially folded a-chains bind calnexin (p88) until B2-microglobulin binds. - MHC complex released from calnexin, binds chaperone proteins (calreticulin & tapasin) & to TAP-1 - antigens processed + delivered via TAP to MHC-I mol - MHC-I completes folding + released from TAp complex & exported
58
Processing of exogenous antigen to MHC-II
- antigen endocytosed, froms endolysosome - antigen degraded by lyosozymes - MHC synthesised in rER w/ associated peptide-invariant chain - MHC fuses w/ endolysosome + invariant chain degraded & replaced by antigenic peptides - MHC/peptide exported to cell surface -> presentation
59
Function of the invariant chain in MHC-II presentation
Prevents other proteins in ER from binding MHC-II, reducing autoimmune risk. Cleaved in acidified endosome, leaving CLIP still bound blocking peptide binding. HLA-DM binds MHC-II & releases CLIP -> MHC-II binds antigen & exported
60
What happens once antigen bound to MHC?
TCR binds both antigen & MHC -> specificity determined by both antigenic epitope & MHC structure (a2-a1 or a1-B1) -> MHC restricts recognition to highly specific TCRs
61
What is the function of co-receptors?
Accessory role in strengthening TCR interactions CD4 - recognise antigen on MHC-II (binds B2 domain) CD8- recognise antigen on MHC-I (binds a3 domain) Enhance affinity of TCR for peptide-MHC complexes
62
Advanatges of MHC-associated recognition
- Extra pathogen recognition mechanism - Recognising different parts of pathogen from antibody - Some peptides are from functional parts of protein (eg essential enzyme sequences) - Can detect antigen that is inside cells (ie CD8 T cell detecting Class I MHC-associated peptides) - Less scope for mutations in pathogens to avoid recognition
63
What arethe anatomical requirements for specifc immunity?
- Enable rare antigen-specific lymphocytes to encounter their antigen - Enable different types of antigen specific lymphocytes to make physical contact - Enable cells to go where they are needed (site of infection) -> requires cell movement to AND within tissue/organ
64
Methods of enhancing rare antigen specifc lymphocyte interactions
- collect lymphocytes/immune cells at sites likely to be infected - circulation & specialised sites of antigen capture to enable interaction
65
What is the lymphatic system composed of?
Primary tissues - lymphocytes develop, Ag independent (thymus, bone marrow) Secondary tissues - lymphocytes migrate to (lymph nodes, spleen, MALT) Lymphoid vessels connect tissues w/ lymph nodes + bloodstream - lymph fluid, lymphocytes + tissue dendritic cells - no pump pressure by muscles
66
Afferent vs Efferent lymphatic vessels
Efferent - exit lymph nodes (away from node) Afferent - supplies lymph nodes (towards node) Drainage lymphoid system asspciated w/ most internal organs - BUT enriched at barrier mucosal surfaces e.g. adenoids + tonsils, Peyer's patches
67
Where dooes lymphocyte activation take place?
Dendritic cells travel to lymph nodes via afferent. T & B cells activated (selection, differentiation + proliferation). Activated lymphocytes leave nodes via efferent lymphatic vessels to blood, then back to tissue
68
True or false: circulation of lymphocytes is periodic.
FALSE - its constant-> increases chance of interaction in peripheral lymph nodes. Can take 4-6 days if antigen is new to the system.
69
Describe 4 key regions of the lymph node structure
Highly spatially organised Cortex - B cell area contsining follicles Paracortex - T cell area Medulla - plasma cells, some T cells + macrophages Secondary follicle - site of intense, transient B cell proliferation during infection
70
What is MALT and dive other types?
Mucosal associated lymphoid tissue - has as many lymphocytes as rest of body. Also gut (G), nasal (N), bronchus (B) & skin (S) asscoiated Also found around specialised cells e.g. liver + reproductive tract
71
Describe Peyer's patch structure ## Footnote GALT
Sub-epithelial dome with no afferent lymphatics or APCs/lymphocytes draining in. M cells in epithelium allow direct antigen entry from gut to GALT. Activated cells move out of germinal centres via efferent lymphatics - drain to blood via thoracic duct.
72
How do M cells on GALT interact with immune cells?
Wrapped around immune cells - close interaction of gut lumen + dendritic/immune cells, movement across detected quickly.
73
Role of the spleen
Filters antigens from blood (no afferent lymphatics) - enables response toblood-borne pathogens. Blood -> splenic artery -> trabecular arteries -> central arteries -> arterioles -> red pulp -> periarteriole lymphoid sheath (PALS) Red pulp deals w/ old RBCs. White pulp areas -> immune function (has B cell follicle germinal centres & PALS) ## Footnote PALS - T cell activation area
74
What is the function high endothelial venules?
HEV let lymphocytes leave bloodstream & enter lymph node
75
What 2 molecules control lymphocyte migration?
Adhesion molecules (organ region) Chemokines (specific location)
76
What are the steps of migration?
Selectin mediated adhesion Firm attachment Extravasation
77
Describe selectin driven adhesion in migration
- P selectin induced on vessels in response to e.g. Lueokotriene B4, C5a, histamine, TNFa, LPS - TNF & LPS also induce additional 2nd selectin (E-selectin) few hours after - Selectins recognised by sulphonated sialyl-Lewis X structure on immune cells -> mediates reversible movement /"rolling" (slows down)
78
Describe firm attachment of immune cells to endothelium in migration
- ICAM-1 + ICAM-2 induced on vessels by e.g. TNF-a (Nf-KB, ROS signalling) - Bind to LFA-1 & CR3 on leukocytes - Rolling stopped due to firm attachment of lymphocyte to blood vessel
79
Deescribe extravasation of immune cells
- Leukocyte crosses endothelial wall, involves LFA-1/CR3 - CD3 interactions also facilitate this (on leukocyte + endothelial cells) - Movement also known as diapedesis
80
What adhesion molecules can be used for lymphocyte migration?
- L-selectin (all types of leukocyte) binds CD34 - LFA-1 integrin (T cells, monocytes, macrophages + dendritic cells) binds ICAM-1, 2-3 - VLA-4 binds VCAM-1 & vibronectin - CR3 binds ICAM-1 - PSGL-1 binds E- and P-selectin
81
Role of chemokines in migration
Cytokines that control movement of cells CXCL8 (neutrophils) & CCL2 (monocytes + NK cells) produced at site of infection. Immune cells can move up chemokine gradient towards infectious pathogen. They draw in cells + also involved in activation/survival of those cells ## Footnote BUT broad chemokine range allows specific localisation of lymphocytes in lymph nodes.
82
Where do CXCL8 & CCL2 act?
CXCL8 - released by macrophages, attract neutrophils + mobilises naive T cells CCL2 - produced by Ep cells + stromal cells -> attracts monocytes
83
Where are inactivated lymphcytes located?
Lymph nodes to allow response to antigen. Dependent on L-selectin (CD62L) expression on naive T cells + P- & E-sleectin on endothelium. - occurs via HEV expression
84
What cells make antibodies? Describe their structure
Plasma cells differentiated from anitgen specific B cells. Have heterochromatic nucleus, lots of golgi + rER for Ab production.
85
Summarise B cell development
They rearrange Ig genes in bone marrow - independent of antigen but needs stromal cells. Rearranged Ig expressed on mem as IgM - eliminated if self-reactive. Cells then mature -> begin to express IgD Mature B cells that leave bone marrow cannot secrete Ab
86
What are the requirements for B cell activation?
B cells require 2 signals to become activated (like T cells) - Signal 1 is recognition by antigen specific mem Ig mol - Signal 2 is often from an interacting CD4+ T cell (T cell dependent stimulation) ## Footnote CD40 (B) interacts w/ CD40L (T)
87
T cell independent B cell stimulation
Bacterial polysaccahrides can deliver strong enough antige to activate B cells -> many antigens close together in repetetive mem No affinity maturation SO Ab not as strong or efficient
88
What happens to B cells in the lymph node when antigen enters?
B cells in cortex. Drained antigens collected by macrophages in marginal zone of cortex -> allow conformational antigen recognition/presentation (no processing) > 1 B cell must be stimulated -> cross-linking
89
What is the role of primary foci?
Region of B/T cell proliferation at cortex/paracrotex border in medullary cords. Some cells then migrate to primary follicle -> form germinal centres
90
T cell dependent B cell activation
B cell can express MHC-II -> process/present antigen. Specific Tfh can bind peptide deliver 2nd activation signal: TCR:BCR, CD40L:CD40, cytokine delivery (IL-4)
91
How can Th cells cirectly deliver IL-4 to B cells?
T cell adheres to B cell+ synthesises IL-4 & CD40 ligand. Th reorients cytoskeleton & secretory apparatus towards B cell. IL-4 released + confined to intercellular space -> taken up by B cells -> promotes proliferation at germinal centres, Ab isotype switching to IgG1 & IgE
92
What is the 1st type of Ab produced against antigens?
Differentiated plasma cells secrete IgM - low affinity. B cells can undergo class switching e.g. reguated by IL-4, IL-5, IFN-y, TGF-B
93
How and why do B/Tfh cell complexes move to primary follicles?
Follicular dendritic cells (FDCs) form network in primary follicles - not DCs (not haematopoietic) Designed to hold Ag/Ab complexes on icosomes on their surface. FDCs hold antigen for extended period, Ab internalised by B cells -> germinal centre formation ## Footnote B cells proliferate rapidly in primary follicles.
94
Formation and structure of the germinal centre
B cells enter primary follicles + down regulate Ig receptors + proliferate extensively -> centroblasts Centroblasts (dark) - B cells expressing IgM Centrocytes (basal light) - proliferative Secondary B blasts (apical light) - differentiation of plasma + memory cells ## Footnote Surrounded by mantle zone - displaced mature B cells
95
What process occurs early in affinity maturation
- Centroblasts undergo hypermutation of H & L chains of their specific Ig molecule - This causes changes in structure of hypervariable regions of Ab, random process
96
How are cells selected to produce high affinity Ab? ## Footnote Affinity maturation
- Centroblasts now stop dividing and re-express their surface Ig – now called centrocytes - move over the FDCs expressing antigen. If they bind antigen with high enough affinity they will receive a survival signal – if not they die!!! - Only the cells capable of secreting high affinity antibody survive - As immune response progresses, feweer Ag means increased competition -> highest affinity selected for ## Footnote Affinity increase of x10-100,000
97
What is class switching?
B cell can change H chain constant region from u (IgM) to gamma (IgG), alpha (IgA) or epsilon (IgE). Variable regions remain constant so no change to affinity. ## Footnote Occurs during centroblast/centrocyte stage
98
What is class switiching controlled by?
CD4+ Th cells & cytokines. W/o CD40-CD40L -> only make IgM Cytokines can also ifluence quantity of Ab made.
99
Role of specific cytokines in Ab class switching
IL-4 - induces IgG1 + IgE, inhibits IgM+ IgG3 + IgG2a IL-5 augments IgA production IFN-y - induces IgG3 + IgG2a, inhibits IgM + IgG1 + IgE TGF-B - induces IgG2b + IgA, inhibits IgM, IgG3
100
What combinations of cytokines can promote class switching?
IL-21 + IL-4 -> IgG1 IL-13 + IL-4 -> IgE IL-10, IL-21 + TGF-B -> IgA
101
Properties & function of memory cells
Survive long periods, undergone class switch + affinity maturation, very fast/efficient secondary response after repeated antigen detected. e.g. IgE binds mast cells to clear worm infection
102
Properties & function of plasma cells
Can be short or long lived. Some stay in nodes/spleen + secrete Ab for few weeks, others go to bone marrow + secrete for many months. - bone marrow main Ab source Very high secretion rate (>2000 molds per second)
103
Production of memory/plasma cells
Production of both occurs in apical light zone of germinal centre - If B cell interacts w/ CD4+ Tfh cell, binding of CD154 on Tfh cell to CD40 on B cell turns into memory B cell -> can leave lymph nodes + enter circulation ## Footnote Dont understand what determines phenotype.
104
Differences between genetic encoding of innate vs adaptive immunity
Innate - germline encoded (inherited), pattern recognition receptors fro PAMPS, MAMPS + DAMPS Adaptive - somatically generated (multiple gene segments rearranged in limited number of genes in developing T/B cells) - highly diverse random reprtoire of potential specificities -> recognise more epitopes
105
Clonal selection of naive lymphocytes
Progeitors -> naive circulating lymphocytes (unique specificity) Upon useful epitope recognition, useful clone proliferated to combat threat. - memory cells also generated
106
Effector mechanisms of Abs vs T cells
Ab: recuited by FcR interactions of different soluble Ab isotypes T cells: require cell contact (MHC processing), or local secretion of cytokines
107
What did sequencing of Ab variable regions discover?
3 hypervariable regions (HV1-3) w/ matched end loop regions - can tolerate variability w/o structural damage HV regions form tight B-barrel -? antigen binding site
108
What unique features to TCR & antibody genes have?
e.g. k-light chain gene (constant region) V and J (joining) segments undergo somatic recombination - different expression in B cells (due to gene rearrangement) -> clonal B cells w/ variable light chains, different binding properties CRD1+2 encoded by Vk, CDR3 encoded by Jk - recombination determines segments included.
109
How is heavy chain gene arrangeent different to light chains?
Has another segment (D - diversity) 2 rearrangements occur sequentially - D->J, V-> DJ, VDJ = V exon, V D & J all contribute to CDR3 Imprecise joining adds more variability to recombination
110
What are recombination sigal sequences (RSSs) for?
Made of conserved heptamer, spacer (12/23) & a nonamer They flank V, D & J segments - link them together during recombination. Spacer is 12 or 23 base pairs, 12/23 rule -> 12 must bind 23, vice versa. SO Segments pulled together + RAG1/2 cleaves within heptamer, intervening sequences excised.
111
Molecular mechanism of V(D) J recombination
RAG1 + 2 turned on, segments pulled together + cleaved above heptamer region. - interveing sequences circularise (comp signal ends) + leave. Coding ends (hairpin loops) on segmenets recognised as damaged DNA by ubiquitous repair machinery -> joins DNA together - NHEJ using DNA-PK, XRCC4, LIGIV, Artemis enzymes Lymphoid specific TdT adds in random nucleotides to joined segments.
112
How does imprecise joining of segments generate junctional diversity?
- RAGs bind + cleave RSSs -> hairpin coding ends - resolution of hairpin -> palindromic P-nucleotides - N-nucleotides added by TdT - pairing of strands + exonuclease cleavage followed by DNA synthesis + ligation -> coding joints Imprecision increases chance of random addition - frameshift/premature stop codon -> non-productive joins
113
What are 2 distinct sources of diversity in the primary antibody repertoire?
Combinatorial - multiple gene segments, any H w/ any L chain Junctional - P nucleotide & N-nucleotide addition, exonuclease activity - largely localised to CDR3 joins between V(light)/D(heavy) & J
114
Segment recombination in TCRs. Compare to BCRs.
TCR has a & B chain loci - segments also flanked by RSSs recognised by same specific RAG proteins B & T receptors similar gene organisation + same recombination machinery. Gene rearrangement take splace in bone marrow + thymus. - many more J segments in TCRs (13B + 61a) - D segments read in all frames, TdT activity at all junctions -> far greater junctional diversity in TCRs (2x10^11) vs Ig (3x10^7) ## Footnote TCR diversity focused on CDR3 - larger contact to antigenic peptide on MHC
115
How are TCR & BCR gene rearrnagements regulated?
Via series of checkpoints linked to lymphocyte development -> ensures allelic exclusion + clonality
116
Describe stepwise regulation of BCR gene rearrangement
H chain: Early-pro -> D-J rearrangements Late-pro -> V-DJ rearrangement on 1st chrom (can be on 2nd chrom as backup if required - cell lost otherwise) L chain: Pre B-cell -> k gene rearranged on 1st chrom, or 2nd chrom, or delata gene on 1st chrom, or 2nd chrom If kappa rearranged -> u:k IgM expressed If delta rearranged -> u:delta IgM expressed ## Footnote Failed joins are non-productive
117
What happens at the checkpoint after late pro-B cell stage?
Productive H chain (V-DJ) combines w/ surrogate L chain + reaches mem surface (cross-linking of L chain confirms progression). H chains now inaccessible, L chains made accessible
118
Is there more variability in H or L chains?
H because TdT mainly active during H chain joning | TdT - Terminal deoxynucleotidyl transferase
119
How do genetics determine whether IgM is transmembrane or secreted?
Secreted generated by alternative RNA splicing -> becomes soluble Alternative polyA site used altering H-chain C-terminal domain SO Pol-III runs through until stop codon at end of SC domain. Caused by upregulation of CstF64 (binds GU rich sequences) + CstF complex -> selection of proximal polyA site vs distal one | SC is secretory component which pentameric IgM can bind ## Footnote Reversible/regulated
120
Tru or False: Only 1 Ab isotype can be expressed at any time
False - IgM & IgD co-expression regulated by alternative RNA splicing -> C-u vs C-delta expression ## Footnote Reversible/regulated
121
Primary Ab vs secondary Ab repertoire
Primary is IgM in bone marrow. Secondary - undergone somatic hyermutation (affinity maturation, mutated V) and class switching (functional skewing, different C) - mutation/selection cycles in germinal centres improve response over time
122
Dendritic cells
Dis by Steinman 1973 - best APC for activating/programming naive T cells Have MHC + extra stimulatory signal (CD80, CD86) - long finger like processes (dendrites) - Immature DC migrate from bone marrow via blood to enter tissues - In tissues they sample env + act as sensory for damage or infection - Very motile
123
What are the 4 main types of dendritic cells?
Classical/conventional DCs (cDCs) Plasmacytoid DCs Monocyte derived DCs (inflam) Langerhans cells
124
Classical or Conventional DCs (cDC)
Tissue resident + have cDC1 & cDC2 subsets Diverse family - CD8a (lymphoid), CD11b (myeloid) & CD103 (enriched in gut)
125
Plasmacytoid DCs
- Sentinels for viral infections - Secrete large amounts class I interferon - Express intracellular PRR TLR7 and TLR9 (eg viral recognition) - Less effective at priming naïve T cells
126
Monocyte derived DCs
- monocyte derived - M-CSF and CCR2 dependent - Variety of roles e.g. supporting effector T cells ## Footnote Inflammatory M-CSF binds receptor called CSF1R, which is a tyrosine kinase that triggers signaling pathways that lead to the growth and differentiation of cells
127
Langerhans cells
- In skin - More similar to macrophages but some functional overlap with cDC - Self-renew in tissue
128
cDC1
BATF3 or IRF8 dependent - Good cross-presentation to CD8+ T cells - Express CD8aa, DEC-205 or CD103 - Produce more IL-12 than cDC2s - Express toll-like receptors (TLR3) - viral recognition ## Footnote IL-12 heterodimer inducing Th1 deifferentiation & IFNy production
129
cDC2
IRF4 dependent - Prime naïve T cells - Associated w/ variety of CD4+ Th cell responses - Express CD11b, DC immunoreceptor (DCIR2), CD301b, CD4 or signal regulatory protein-a (SIRPa) - Difficult to distinguish from MoDCs + other DCs induced during inflammation ## Footnote SIRPa binds CD47 (ubiquitous), DC survival, migration, and phagocytosis.
130
What is the unique function of intestinal DCs?
CD103+ DCs in gut drive tolerance to oral antigens from food + commensal bacteria. - induce Treg generation (anti-inflam) - dependent on TGF-B & retinoic acid (vitA mtabolite)
131
Describe features of cDCs in normal tissues
Immature w/ many dendrites - Ag capture/migration Low CD80/86 expression, but have CCR5+6 chemokines. They endocytose soluble Ags - many endosytic vesicles (MHC-II + lysosomal proteins)
132
Which MHC mols process extra- vs intracellular antigens?
MHC-I: intracellular MHC-II: extracellular
133
What are the different routes of immature DC antigen capture?
- Receptor mediated - Macro-pinocytosis - Viral infection - Cross-presentation after phagocytic/macropinocytic uptake - Transfer between DCs
134
What does cross presentation allow?
Exogenous proteins presented by MHC-I, enabling viral recognition w/o DC infection - Peptides taken up from pathogens, infected dying cells + tumour cells - Taken up via MHC-II then processed to be presented via MHC-I
135
How are immature DCs in peripheral tissues activated?
By PAMPs on pathogens. Receptors include: - Pattern recognition receptors e.g. TLRs - Some phagocytic receptors also activate DCs: ○ DC-SIGN recognise mannose, fucose on pathogens ○ Dectin-1 recognise B1,3 glucans in fungal cell walls ○ Receptors for complement
136
DC maturation
Licenced by pattern recognition receptors. - TLR7/8 signalling induces CCR7 & enhances antigen processing - CCR7 dircets migration to lymphoid tissues + augments co-stim & MHC mol expression - mature DC in paracortex can then prime naive T cells ## Footnote DAMPs (uric acid) or cytokines can also promote licencing
137
How do DCs change when licenced (matured)?
TLR signalling -> induces CCR7 receptor for CCL21. Mature DCs migrate via lymph to lymphoid tissues: mem folds when in circulation, express lots of stable MHC when in lymph nodes High costim mols (CD80/86), stop taking up antigens (poorly endocytic). - also attract naive T cells (express adhesion mols + secrete CCL18) ## Footnote adapted to talk to T cells
138
Function of mature DCs
- Present Ag to naïve T-lymphocytes, priming - Activate any antigen specific T cells to divide into effector cells that enter circulation
139
How do DCs polarise an immune response?
Different subtypes: cDC1 (IRF8+) - Th1/CD8, cDC2 (IRF4+) - Th2 Maturation factors: PAMPs, peptidoglycan promotes IL-12 (Th1 immunity), histamine promotes Th2 (suppresses IL-12) Accessory cells: ILC2, eosinophils support Th2 (IL-5, IL-13 produced) Cytokines: IL-10 & TGF-B promote treg generation
140
Which cytokines and TFs induce Th1 & Th2 response?
Th1: type 1 PAMPs -> trigger IFN-y, IFN-a, IL-18, CCR5L - IL-12 + 27 for co-activation T cells Th2: type 2 PAMPs -> histamine, PGEp, TSLP & CCR2L ## Footnote Th1 produce IFN-y & TNF-B Th2 produce IL-4 & IL-5
141
Are DCs or macrophages better APCs?
DCs Macrophages mainly interact w/ already primed effector CD4+ T cells. DCs present processed antigen + drive clonal expansion/differentiation of T cells into effector cells.
142
How can DCs be trained to kill tumours?
Monocytes cultured + differentiate to DCs. DCs cultured ex-vivo with elctroproated tumour lysate proteins OR synthesised peptides. Infusion into individual. ## Footnote Both host + tumour cell dterminants need to be considered -> highly heterogenous
143
Compare different types of DC vaccines
1st gen: immature patient isolated or ex-vivo generated mo-DCs, loaded w/ synthetic antigen/tumour cell lysates - limited success BUT safe e.g. Sipuleucel-T 2nd gen: matured mo-DCs, more refined antigen prep methods, better clinically e.g ACAM2000 Next gen: focus on specifc subsets + naturally occurring DCs -> initial trials safe + more effective
144
What co-stimulatory molecules provide 2nd signal for activation of T cells?
CD28 on T cells. CD80/86 on DCs - presence can be upregulated by pathogen presence
145
What happens if there is no 2nd activation signal for T cells?
Become anergic - unresponsive to antigen Can be useful mechanism to prevent-self-reactivity.
146
How do cells adhere to each other during T cell scanning and activation?
LFA-1 & ICAM-1 interaction -> weak adhesion between CD4+ T cell & DC, close contact w/ presented antigens If correct antigen found - TCR complex w/ MHC-II -> signalling increases affinity binding of LFA1-ICAM1 interaction (conformational change) If correct antigen not found - tethering not strenghtened, T cell moves to look for DC w/ recognisable antigen
147
What are the specific contacts between CD4+ T cells & DCs called?
Immunological synapse has specifc conformation. Made up of cluster of mols at T cell/DC interface -> supramolecular activation clusters (SMACs)
148
SMAC structure for T cell/DC interaction
Outer ring (pSMAC) - LFA1:ICAM1, larger mols so physically excluded from cSMAC Inner circle (cSMAC) - TCR, CD4+, CD28+, MHC:peptide -> high spatial organisation, mols pulled tighter at specific locations between T cells/DC
149
Signalling of T cell activation after antigen binding
TCR no cpaability to sigan downstream SO interacts w/ CD3 complex (w/ intracellular domain/ITAMs) Signals also triggered directly by CD28 co-stimulatory mol. -> activation of PTK cascades + calcium signalling -> alters gene transcription via activation of TFs ## Footnote PTKs - Lck, ZAP-70 Ca2+ signalling - calcineurin (phsophatase) activated -> dephosphorylates NFAT ( nuclear factor of activated T cells), which translocates to the nucleus to regulate gene expression.
150
What happens when CD4+ T cells are activated?
Driven into cell cycle -> clonal expansion of Ag specific T cells Also express IL-2R on membranes + secrete IL-2 (GF) - act in autocrine or paracrine manner (drives proliferation) Differentiation into effector cells ## Footnote Within 3-4 days, 10,000-100,000 fold increase in no. antigen specific CD4+ T cells. ALL occurs in lymph nodes or spleen.
151
Role of cytokines in the variation of immune response
Small protein released by cells that has specific effects on interactions between cells, on communications between cells or on the behaviour of cells. - acts on cell receptors - can have systemic effects, but mostly paracrine effect ## Footnote Different combinations in time + space deliver different messages.
152
Role of Th1 subsets
Respond intracellular pathogens (viruses + some bacteria) - produce IL-2, IFN-y, lymphotoxin (LT) - activate macrophages, stimulate CD8+ T cells to kill infected cells
153
Role of Th2 subsets
Respond extracellular pathogens (helminth parasitic worms) - produce IL-4, 5, 9 & 13 - attract + activate eosinophils/basophils, help promoter tissue repair
154
What do cells require to differentiate into Th cells?
3rd signal - provided by cytokines Signal 3 for Th1 cells: IL-12 from DCs or IFN-gamma from Th1 Signal 3 for Th2 cells: only needs IL-4
155
How are Th subsets regulated?
Th1 & Th2 cross-regulate each other. - Th1 downregulates Th2 by IFN-gamma secretion - Th2 downregulates Th1 by IL-4 secretion Responses become polarised towards dominant response - specialised to pathogen
156
What are newer subsets of effector CD4+ T cell subsets? ## Footnote Discovered past 20 years.
Th17 cells T-follicular helper cells (Tfh) Tregs - reguatory subset
157
What cytokines drive Th17 differentiation?
TGF-B IL-6 IL-1
158
What cytokines are produced by Th17 cells?
IL-17, 21 & 22 Immune responses at epithelial surfaces at lung + intestine - important for fungal/bacterial infections
159
Tfh cells function
Specialised to go to B cell follicles (germinal centres) of lymph nodes to help B cells make antibody via cytokine production. Acquire special receptor (CXCR5) on surface to move there
160
Tregs function
Select against potentially self-harmful T cells in thymus. Subset of high affinity T cells differentiate to Treg phenotype instead of deletion. - produce reg cytokines to switch off self-reactive T cells -> IL-10, TGFβ, and IL-35 (immunomodulatory) ## Footnote supress self-harmful immune responses, autoimmune patients often have dysfunctional Tregs
161
What marker was identified for suppressive CD4+ T cell subset (Tregs)?
CD25 Foxp3 also identified as critical Treg gene - key determinant of autoimmunity.
162
2 methods of treg formation
Produced in thymus as normal part of T cell development OR Made in peripheral tissues/organs - induced by IL-2/TGF-B 3rd signal -> induced/peripheral Tregs
163
Treg immunomodulatory functions
TGF-B & IL-10 suppress immune cell expression. Tregs also for: - Anti-inflam cytokines - Outcompete effector T cells for resources - Kill self-reactive T-cells
164
What is the result of a lack of Tregs?
Autoimmune disease - Foxp3- mutant in mice -> scurfy animals dont grow In humans -> IPEX, X-linked only in boys, many autoimmune diseases associated
165
T cell plasticity
CD4+ subsets able to become other subsets - not solely one-way traffic.
166
Which myeloid cells are phagocytes?
Macrophages + neutrophils: kill pathogen Dendritic cells: present pathogen ## Footnote Macrophages can act as APC later in immune repsonse
167
Which myeloid cells are granulocytes?
Mast cells, eosinophils, basophils & neutrophils ## Footnote Many innate immune cells ativated by PRRs e.g. TLRs
168
Macrophage location + function
Reside in tissue, produced from either monocytes from bone marrow (enter tissue from circulation) or embryonic cells during development. - sense/move towards pathogens via chemotaxis - phagocytic receptors on membrane
169
Which chemokines direct chemotaxis of macrophages?
CCL8, 9 + 21 receptors mediate M1 chemotaxis CCL7 receptors mediate both M1 & M2 chemotaxis -> polarisation of macrophage responses
170
Localisation + function of neutrophils
90% circulating granulocytes, readily activated, move into sites of inflammation. Short life - pus = dead neutrophils, highly phagocytic. - sense + move towards pathogens via chemotaxis (CXCL8-R)
171
How does phagocytosis kill a pathogen?
Pathogen engulfed into phagosome -> acidified (mediated by V-ATPase proton pumps) Fuses w/ lysosome forming phagolysosome (has enzymes, toxic oxygen species) In neutrophils, cytoplasmic granules & anti-microbial peptides fuse with pahgosome
172
Give an example of an anti-microbial peptide in neutrophils
Cathelicidin LL-37
173
Describe eosinophils
Make up 2-5% blood - rare Toxic granules: eosinophil cationic protein, major basic protein Can be activated to degranulate by - complement, Ab, cytokines
174
Describe basophils
Make up 0.2% blood - rare Toxic granules: histamine, prostaglandins (PGD2), heparin, leukotrienes Can be activated to degranulate by - complement, Ab, cytokines ## Footnote PGD2 - suppress IL-12 production, induce Th2 response ALSO chemotaxis & histamine release
175
Describe mast cells
Basophil-like but found in tissue (not blood). Bind IgE (bound to FceRI) on cell surface w/ high affinity at rest. Mast cell activated when IgE binds antigen (multivalent) -> release granules
176
Describe natural killer (NK) cells
Innate cells that produce cytotoxic molecules to kill 'altered' cells -> infected/tumour cells Express activating and inhibitory receptors (inhib-R binds MHC-I) If cell is infected/cancerous -> downregulates MHC-I for NK cell inhib receptors SO NK cells kill abnormal cells
177
True or False: Immune cells often have redundant functions
True - increases ability to fight complicated infections + acts as fail-safe Innate + adaptive immunity exhibit significant cross-regulation/communication largely via cytokine/Ab production
178
What is the complement system?
Major innate immune system - provides early killing of pathogens. Has > 30 different soluble proteins, made in liver as inactive precursors + present in blood & other bodily fluids in inactive form.
179
Structure of complement enzymes
Cleaved into 2 or more parts - activated Smaller fragment may mediate inflam. Larger fragment has 2 sites -> active site & binding site for next protein cascade
180
What are the 3 pathways of complement activation>
Classical Alternative Lectin All generate C3 convertase, which cleaves C3 -> C3b bound to microbial surface + C3a released
181
Describe the classical complement pathway
C1q binds to one pentameric IgM mol in staple form on bacterial surface OR C1q binds at least 2 Ag-bound IgG mols C1q binding to Ig activates C1r -> cleaves & activates serine protease C1s ## Footnote Ab-dependent
182
Function of C1s in classical complement pathway
Cleaves C4 to C4a & C4b (binds microbe) C4b2a is active C3 convertase So cleaves C3 to C3a + C3b which bind to microbe or to convertase itself
183
Role of C3b in anti-pathogenic response
Opsonin - marks pathogens/immune complexes for phagocytosis Downstream also forms C5 covertase complex (C4b2a3b) -> membrane attack complex (MAC) formation ## Footnote MAC is pore forming protein complex
184
Describe the alternative complement pathway
Complement binds pathgen surfaces + involves complement factors B & D. C3 spontaneously hydrolysed -> C3(H2O), can bind factor B, then cleaved by D into Ba & Bb C3(H2O)Bb complex is C3 convertase, C3 cleaved -> C3a + C3b ## Footnote Ab dependent
185
Describe the lectin complement pathway
Complement binds mannose binding protein - bound to pathgen surface. Mannose binding lectin (MBL) & ficolins bind to pathogen glycoprotein polysaccarides on surface. -> triggers activation of MBL-asscoiated serine proteases (MASPs) which bind MBL MBL-MASP complex cleaves C4 & C2 -> C3 activation ## Footnote Ab independent
186
How does cleavage during complement lead to pathogen removal?
1. Induction of lysis 2. Opsonisation of pathogen 3. Induction of chemotaxis & inflammation 4. Immune complex clearance (small Ab-soluble antigen complexes, not enough Ab to mediate phagocytosis)
187
What is the major function of CD4+ Th subsets?
Cytokine production
188
Main Th1 immune function ## Footnote Cd4+
Normally against intracellular pathogens. Central activators of macrophages via IFN-gamma secretion & CD40 ligand-CD40 interaction. - activated macrophages upregulate MHC-II (more T cell activation) ## Footnote Takes time - prolonged attachment of Th1 to macrophage -> delayed type hypersensitivity (DTH)
189
Other Th1 function ## Footnote CD4+
- induce macrophage differentiation in the bone marrow via IL-3 & GM-CSF - Induce T-cell proliferation, increase numbers of effector cells via IL-2 - Kills chronically infected cells releasing bacteria to be destroyed via Fas ligand & IFN/TNF
190
Main Th2 immune function ## Footnote CD4+
Deal w/ large extracellular pathogens (worms) Can help B cells produce Ab via IL-4 (B cell GF), also function of Tfh. Activate basophils, eosinophils + mast cells via IL-4, 5 & 13
191
Th2 function for macrophages ## Footnote CD4+
Can activate 'alternatively activated' M2 macrophages via IL-4 + 13. These macrophages -> TGF-B + RELM-a, promotes wound healing ## Footnote RELM-a activates fibroblasts to upregulate lysyl hydroxylase 2 -> boost collagen cross linking
192
Th17 main immune function ## Footnote CD4+
Regulate responses against extracellular bacteria + fungi. Produce IL-17 & IL-22: IL-17: act on many cells to induce cytokine/chemokine secretion, neutrophil recruitment, stimulate neut + macroph production in bone marrow IL-22: antimicrobial peptide production e.g. Reg3 family AMPS, lipocalin-2 (sequesters iron to reduce bacterial growth)
193
What is the role of CD8+ T cells in immunity?
MHC-I activated to form CTLs - directly kills. Proliferate/differentiate via IL-2 secretion + autocrine signalling w/ IL-2R After activation, CTLs do not require co-stimulation to kill cells via cytotoxic granule release.
194
How does CTL kill infected cells?
- binds target losely - recognises antigen (via MHC-I) & Tc cytoskeleton reorganises, granules moved to contact site - cytotoxic granules released at cell-cell contact (specific killing)
195
What is contained within CTL/Tc granules?
Granzymes - serine proteases that induce apoptosis Granulysin - antimicrobial + induces apoptosis Perforin - helps deliver granule contents to target cell by pore formation in cell membrane Directed granule secretion prevent viral spread to other healthy cells.
196
Do CTLs/Tc produce cytokines? ## Footnote CD8+
Yes. - IFN-gamma to activate macrophages, increase MHC-I expression in infected cells (pos feedback) & directly inhibit viral replication - TNF-a/LT-a helps IFN-gamma activate macrophages, helps directly kill infected cells
197
Coordination of CD8+ & CD4+ Th1 cell responses to intracellular pathogens?
Both activate macrophages via IFN-gamma. CTL directly kills infected macrophages. Th1 differentiation increased via IFN-gamma. Also stimulates T cell proliferation (-> CTLs) via IL-2 secretion.
198
What are the effector mechanisms of antibodies?
Not directly toxic, but facilitate immune mechanisms. Neutralisation Agglutiniation Opsonisation ADCC Classical complement activation Specialised responses
199
Agglutination ## Footnote Ab mechanism
Clumping of particles -> latice work more likely to be phagocytosed Can involve complement (removing immune complexes)
200
Opsonisation ## Footnote Ab mechanism
Complement & IgG coat bacterium. C3b binds CR1 on macrophage & conserved IgG region binds Fc receptor -> phagocytosis
201
Ab dependent cell mediated cytotoxicity ## Footnote Ab mechanism
Ab binds infected cell or large extracellular pathogen Ab makes bridge to immune cells via FcR + mediates death via degranulation/innate directed killing
202
Specialised immune responses of antibodies
IgE-mediated activation of mast cells -> degranulation SO lipid mediators, cytokines, granules (e.g. histamine) are released Initiates inflammatory response (increased vasodilation, increased vascular permeability ->accumulation of immune cells at site of infection)
203
Monoclonal vs polyclonal antibodies
Monoclonal antibodies are produced by a single B cell clone and bind to a single epitope on an antigen, offering high specificity and consistency. Polyclonal antibodies, on the other hand, are produced by multiple B cell clones and bind to multiple epitopes on the same antigen, providing broader recognition and sensitivity
204
Making polyclonal Abs
Antigen samples injected, immuised many times over 10 weeks -> maximise class switching + selection Serum enriched for specifc Abs against injected antigen. - used for diptheria 'anti-toxin' 1890 - convalescent plasms confers passive immunity to recipients (high Ab levels)
205
What is a hybridoma?
Used to make mAb. Has Ab producing gene from B cell fused w/ myeloma that has immortal + highly proliferative properties.
206
# [](http://) How are hybridomas and then mAbs produced?
Mouse immunised repeatedly w/ antigen -> class switching + increase affinity. Spleen cells removed containing antigen-specific B cells (splenocytes) - isolated. - splenocytes fused in vitro w/ myeloma - myeloma has purine enzyme deficiency + cannot secrete Ig - cells cultured in HAT medium, only fused cells that can utilise purine survive Then plated so 1 hybridoma per well. Hybridoma Abs screened agaisbt orignal antigen + clones selected -> immortal lines secreting mAbs | HAT - hypoxanthine, aminopterin & thymidine
207
What are some advantages of mAbs?
- High specificity - Low cross-reactivity - Standardised worldwide - Unlimited supply - Many different uses i.e. immunoassays, diagnosis, tissue typing
208
What is a disadvantage of antibodies?
In vivo - trigger immune response but not access sites needed -> off-target effect SO can create Fab fragment delivery system using insects to circumvent immunity.
209
What are some applications of mAbs?
- affinity chromatography - immunoisolation (magnetic separation) - (haem)agglutination - ELISA/ELISpot - Immunohistochemistry/fluorescence - Western blotting - Flow cytometry
210
mAbs for affinity chromatography
mAb bound to beads used to purify Ag from a mixture of molecules Unbound mols washed away + purified antigen eluted
211
Immuno-isolation ## Footnote Magnetic separation of cell populations
Obtains purified cell populations, can be sed clinically. mAbs coupled to paramagnetic beads + poured over iron wool mesh -> magnetic field applied Cells specific to Ab stick to mesh + othrs washed out
212
mAbs for agglutination
Identify if Ab present for an Ag + visualise clumping as complexes form - used in blood typing + test if patirnt had infection (serums mixed w/ pathogen culture Haemagglutination to test for blood type: A has anti-B Abs, B has anti-A Abs. Abs colourless so need fluorescnet markers (FITC), metal ions or enzymes markers (peroxidase)
213
mAbs for ELISA
Used clinically for viral diagnostics, hormone levels etc. mAb added, covalently linked to enzyme. Susbtrate added which binds enzyme to induce colour change. - measure using absorbance Secondary Ab can be used for increases sensitivity - ammplifies signal
214
mAbs for ELISpot ## Footnote enzyme linked immunospot
Highly sensitive assay - measure freq of cytokine/Ab secreting cells at single cell level. Cells cultured on +/- stimuli w/ capture mAb, cells removed but Ab remains on plate - secreted Ab then visualised using substrate reaction: - detection mAb-biotin added w/ streptavidin enzyme + ALP for precipitation | ALP - alkaline phosphatase
215
mAbs for immunohistochemistry/fluorescence
Shows localisation for Ag - can be quantitative. Needs fresh, frozen, fixed tissues. - useful for DNA seqs on chroms, spatial/temporal patterning of gene expression Histochemistry - uses enzyme substrate reactions -> anti-glucagon & anti-insulin Abs
216
mAbs for Western Blotting
Qualitative + quantitative analysis of protein expression -> tissue homogenates or cultured cells. Can be used diagnostically (Lyme disease, HIV) SDS-PAGE before transfer over to nitrocellulose membrane w/ antiserum. Enzyme linked anti-IgG used to detect bound Ab (secondary)
217
mAbs for flow cytometry
Abs against different cell durface proteins - lasers detect fluorescent Abs bound to cells. -> qualitative & qunatitative info : level of activation, cell viability, cytokine secretion, rare cell types Used clinically - tumour detection, monitoing cell number ## Footnote produces scatter plots
218
Therapeutic uses of mAbs
TNF therapy - mAbs for TNF-a used for RA, IBD, psoriasis & ankylosing spondylitis Can also be used to enhance T cell responses by blockaing immune checkpoints. - anti-PD1 & anti-CTLA-4 Abs -> Can boost immune responses to tumours
219
What are the 2 features of immunological memory?
Primary adaptive response to infection often slow & weak -> protective immunity Secondary exposure induces greater, more rapid response w/ higher affinity. -> memory
220
What immune cell changes are associated with memory?
- Expansion of 'clones' of cells w/ re-arranged antigen receptor genes specific for primary antigen encountered * TCR * BCR/Ig - Enhanced migration & re-stimulation properties: adhesion mols, rapid effector function - Survival: maintenance of clones, responsive to growth/cytokine survival signals
221
Features of memory B cells
Ag specific, already undergone class switiching + affinity mat. Can re-enter germinal centres in secondary response -> somatic hypermutation + affinity mat BUT needs CD4+ Th cell help for Ab secretion (not a plasma cell)
222
Which cytokines mediate memory T cell survival & maintenance?
IL-7 & IL-15 -> homeostatic proliferation
223
What are some features of memory T cells?
- Express high cytokine levels - IL7R for survival & genes like bcl2 - High adhesion molecules - Lower expression markers associated w/ effector cells e.g. CD69 Can be tissue resident or adapted for recirculation through lymphoid (central) or adapted to rapidly re-enter inflamed tissues (effector)
224
Memory vs naive b cells
- mem also have IgG, IgA & IgE - mem have high Ig complement receptor - mem in bone marrow, lymph node + spleen but naive only in spleen - both recirculate - mem higher affinity - mem higher ICAM-1 expression
225
History of immunisation
Variolation portected against smallpox in 1500s - in UK 1717 by Lady Montagu BUT 1% death rate Jenner 1796 1st small pox vaccine from cowpox (innoculation) Pasteur vaccines for rabies + anthrax (1879), based on weakened pathogen forms
226
How do modern vaccines work & what do they require?
Generate protective antibodies & inducing T cell & B cell memory. Requires Ags from targte pathogen & infection signal (alert immune system) - Live & attenuated naturally provide both - Other vaccines provide antigens but need adjuvant to provide infection signal - Some need boosters
227
Name the 6 main vaccine designs in widespread use
Live attenuated Killed/inactivated Portein subunit Recombinant viral-vectored Virus like Nucleic acid based (mRNA)
228
Live attenuated vaccines
Related, less harmful infection: * Vaccinia (cowpox) related to Variola (smallpox) * BCG bacteria (bovine tuberculosis) related to Mycobacterium tb. * Highly effective, rarely found in nature, dangerous to immuno-compromised patients. Live attenuated pathogen: * Sabin oral Polio, MMR, chickenpox * Highly effective, some risk of disease in immuno- compromised patients
229
Killed /inactivated vaccines
* Salk Polio virus * Lower risk of disease but can occur due to improper activation * May need boosters * Inactivated using heat, chemicals (formaldehyde), radiation to preserve antigen structure but stop pathogen replication
230
Protein subunit vaccines
* Use antigens that best stimulate the immune system e.g. Hepatitis B, Novavax vaccine for SARsCOV-2 * May be linked to e.g. boosting mechanism such as a bacterial stimuli e.g. HiB meningitis
231
Recombinant viral-vectored vaccines
* Virus bioengineered to express target pathogen in vivo * Widely investigated w/ good safety e.g. Ebola * Often use non-human virus carrier e.g. simian adenovirus used for AZ SarsCov2 vaccine If use same carrier repeatedly -> causes reactivity to carrier virus
232
Virus-like particle vaccines
* Highly effective, closely resembles live virus but non-infectious * Can have multiple antigenic components e.g. Gardasil for Human Papilloma Virus
233
mRNA vaccines
* Can encode many antigens (even whole genome) * Antigen encoding mRNA complexed w/ a carrier like lipid nanoparticles to be delivered into target cells * Antigen transiently expressed by target cells -> immune response * Non-infectious & quick to manufacture, can be tricky to store as requires cold chain to store e.g. SarsCov2 Moderna & Pfizer
234
What do adjuvants do?
Chemicals that boost immune response to an Ag - stimulate inflam + T cell activation via APC e.g. oil/water emulsion, alum, TLR ligands, liposomes
235
What is a limiting factor of vaccine efficacy and how can this be subverted?
Individual MHC type -> not all Ag immunogenic in everyone + some pathogens mutate rapidly (many life cycles) SO multivalent vaccines deliver variety of Ag epitopes w/ adjuvant -> ranging immune response
236
Mucosal vaccines
Delivered via nose/throat (inhalable/sniffable) More tissue specific - can block respiratory viruses + potentially more durable Used for flu + investigated for COVID-19 ## Footnote e.g. Fluenz Tetra/FluMist
237
Why dont we have vaccines for all diseases?
- Complicated pathogen life cycles - malria has stage specific immune responses - Antigen variability - Latency (HIV/Herpes viridae) e.g. no vaccine for HIV + TB (1/4 global pop infected) Also lack of research into torpical zoonotic infections e.g. Zika, Ebola, COVID-19
238
Which factors contirbute to vaccine hesitancy?
Socioeconomic factors - education, beliefs Social influences - professional recommendations, social media, previous experiences Trust - efficacy & safety of vaccine, misinformation Accessibility - cost, convenience, availability, language barriers
239
Describe 6 themes that contribute to vaccine trust
Control: the idea that we can take back control of our own bodies through alternative means. Parenting style: philosophies that shun mainstream medicine, opting for “natural” remedies. The Past: previous bad healthcare experiences or a bad experience around vaccination shapes perceptions. Risk: misunderstandings about the risks of vaccination versus the risks of disease. Fear of chemicals: the fear that the vaccine will introduce toxic chemicals. Distrust of government agencies and corporations.
240
Herd immunity
Certain prop must be immune to protect the rest of the population from a pathogen - depends on virulence of pathogen + pop susceptibility e.g. measles needs ~90% (very virulent)
241
MMR vaccine misinformation
Wakefield 1998 linked it to autism -> fraudulent + unethical, no replicative study Measles rose in Europe 30-fold
242
How does social media contirbute to vaccine hesitancy?
Misinformation is false or inaccurate information—getting the facts wrong. Disinformation is false information which is deliberately intended to mislead—intentionally making the misstating facts - can be very lucrative Algorithms fuel spread of disinformation further ## Footnote Disinformation dozen: 12 people responsible for almost 2/3 of anti-vaccine content circulating on social media platforms
243
What is tolerance and its 2 broad mechanisms?
Immune system needs broad recognition whilst avoiding self-harm. Self-reactivity normal in healthy immune system BUT restrained by tolerance mechanisms. Tolerance is antigen specific. 1. limit production of self-reactive T & B cell clones 2. prevent unwanted destructive responses by any clones produced
244
Self -tolerance
Failure to respond to intrinsic self-antigens Acquired, not inherited. - Non-identical twin calves sharing common placenta become tolerant of each other - chimerism of blood systems (shared components) - Tolerance may be induced to antigens that are shared as a foetus (able to tolerate skin grafts from each other)
245
Can tolernace to external antigens be developed?
Yes - harmless non-self Ags (commensal bacteria & fetomaternal antigens) - therepeutically relevant Ags e.g. allergens, transplantation
246
Is self-tolerance due to clonal deletion?
NO - protection form autoimmunity is compromise: Removal of dangerous self-reactivity WITHOUT impairing capacity for broad recognition & effective defence. Mechanisms inc: - inactivation of ptoentially self-reactive clones - immune regulation - self-epitopes not available for recognition
247
What are the two types of tolerance?
Central - in primary lymphoid organs (bm, thymus), self-reactive lymphocytes removed during development - not absolute Peripheral - in peripheral organs/tissues & secondary lymphoid organs, many mechanisms to limit self-reactivity
248
Quality checkpoint in B cell central tolerance
Checks ORFs created to make H chain that will fold w/ L chain to form surface receptor (BCR) - monitors VDJ rearrangement If successful, signals cell to downregulate Rag enzymes for recombination -> proliferation before rag genes turned back on ## Footnote Ag independent
249
Tolerance checkpoint in B cell central tolerance
Checks self-reactivity of receptor after VJ rearranging of L chain. IgM now expressed on cell surface B cell fate determined by interaction w/ self-antigen in bone marrow - influenced by conc of antigen & ability to cross-link surface receptors. If self-reactivity dangerous -> clonal deletion OR receptor editing ## Footnote Ag dependent
250
Receptor editing
Repeated VJ rearrangements at L chain foci. If 1st recombination fails, nonproductive join: - rag genes stay on + rearrange again Generates non-autoreactive mature B cell + exits to periphery OR clonal deletion
251
Summarise B cell fates during central tolerance
Non-self reactive -> mature B cell migrates to periphery. Self-reactive clones -> deleted or rescued via receptor editing Self-reactive clones w/ low affinity (non-cross linking) slef mol -> ignorant clones to periphery, cannot bind Ag in enough conc, insufficent context to cause activation Self-reactive clones w/ soluble self-mol - anergic clones to periphery, functionally unrespnsive to Ag stimulation
252
T cell development stages
Lymphoid progenitor ibteracts w/ stroma -> commits to T cell (95% become aB T cells) early thymocyte -> double negative thym (DN) -> double positive thyms (DP) -> single positive thyms (SP) VDJ(beta) recombination at DN stages VJ(alpha) recombination at DP stages ## Footnote Stepwise process DP has both CD4+ & CD8+
253
Role of checkpoints during T cell central tolerance/development ## Footnote In thymus
Checkpoints remove useless/harmful clones: - fail to express TCR - cannot recognise self-MHC mols - recognises self-MHC:self-Ag too strongly ## Footnote Only ~2% of cells that make TCR make it through thymic selection to enter periphery
254
Checkpoint 1 of T cell central tolerance
Has TCR-B gene rearrangement -> functional pre-TCR expression Quality check to see if B-chain can bind to surrogate a-chain + make pre-TCR. - survival signal downregulates Rag - proliferation before a-chain rearrangement Otherwise apoptosed ## Footnote Occurs DN3, Ag independent
255
Checkpoint 2 of T cell central tolerance
Positive selection - can rearranged alpha-beta TCR recognise self-MHC:self-Ag? 10-30% of new TCRs survive + move to medulla regionas SP T cells. IF TCR cannot recognise - rag genes left on + receptor editing of a-chain OR apoptosed ## Footnote Occurs at DP stage.
256
Checkpoint 3 of T cell central tolerance
Negative selection - does alpha beta TCR recognise self-MHC + self-peptide too strongly? DCs + medullary ep cells present Ag to SP thymocytes. Medullary ep cells express AIRE & Fezf2 (transcription regulators) -> allow promiscuous gene expression -> gene mosaicism, provides non-thymus peptides for neg. selection Macrophages clear up dead thymocytes in development.
257
What is T cell selection hypothesised to be based on?
Binding affinity. TCR binding not too strong - T cells survive TCR binding moderate/strong - thymic Tregs (Foxp3+) - useful T cells exit thymus Non-function or overly strong binding -> apoptosis
258
What causes peripheral tolernace in B cells?
Lack of T cell help B cell not activated by Ag so does not differentate to plasma cells In germinal centres: B cell competes w/ follicular DC for Ag. - Ag presentation by DC in absence of danger is tolerogenic (anergic)
259
How does clonal anergy arise?
T cells receive signal 1 alone -> is unresponsive to Ag e.g. when encountered in absence of danger
260
How can pTregs arise?
Driven by presence of Ag & immune suppressive signal 3: Ag + TGF-B -> pTreg (Foxp3+) Ag + IL-10) -> Tr1 (Foxp3-) ## Footnote Immunosuppressive cytokines
261
Function of pTregs & tTregs ## Footnote peripheral vs thymus
tTregs recognise self Ag pTregs recognise self Ag + harmless non-self Ag -> allow temperance of immune responses to harmless non-self Ag (microbiome commensal bacteria/food)
262
How do Tregs regulate immune responses? (inhibit harful T cells)
- Production of anti-inflammatory, pro-regulatory cytokines; TGFβ, IL10, IL35 (dependent on signals driving differentiation of the Treg) - Modulation of dendritic cells - Outcompete effector T cells for resources; IL2 (Treg commonly express IL2R) - Direct killing of T cells - Exosome transfer of miRNAs
263
Are all tissues immunologically euquivalent
NO - gut exposed to many non-self antigens so more tolerogenic.
264
Function of oral tolerance
Allows desensitisation to an antigen via exposure to high tolerance in the gut e.g. ingested poison ivy reduces allergic skin reaction following contact
265
List some immune priveleged sites + describe their features
Antigens in immune privilege sites do not induce immune attack - brain, eye, testis, uterus, hamster cheek pouch - Enclosed by physical barrier - Low MHC-I - Rich in suppressive cytokines (TGF-B) - Express FasL Grafts placed in these sites not rejected - antigens placed at these sites don’t elicit destructive immune response.
266
What causes sympathetic opthalmia?
Immune privilege broken. Trauam at 1 eye -> intraocular Ag acrried to lymph + activate T cells BUT effector T cells return via blood to encounter Ag at BOTH eyes.
267
What mechanisms allow tumours to avoid immune recognition?
Low immunogenicity Tumour treated as self-Ag Antigenic modulation Tumour induced immune suppression - induce Tregs via cytokine secretion Tumour induced privilege sites (TME) Hampers treatments - immunotherpies need to overcome suppression.
268
Autoimmune responses
Immune response againts one or more self-antigens
269
# [](http://) Autoimune disease
Caused by autoimmune response & develops due to failure of self-tolerance. Normal effector mechanisms triggered. - Usually not possible to clear self-antigen -> chronic response 5-8% of pop, >80 different diseases - RA most common
270
Give exmaples of failure in self-tolerance devlopment leading to autoimmunity
AIRE mutants - expression helps screen for storngly reactive T cells Mutations in Foxp3 - linked to IPEX, involved in Tregs development ## Footnote IPEX - immunological polyendocrinopathy X-linked disease
271
What causes IPEX?
Defects in FoxP3 gene (Tregs) -> immunodysregulation, polyendrocrinopathy, enteropathy. - Systemic autoimmunity, begins 1st year life -> diarrhoea, endocrinopathy (diabetes) + eczema Treatments: bone marrow transplant & immunosuppressive agents.
272
Classification of autoimmune disease
Organ-specific: effector function againts Ag confined to affected organ - MS, myelin sheath targeted on CNS axons - Type 1 diabetes, B-cells of pancreas targeted Systemic: widespread Ag found in most cells - Systemic Lupus Erythematosis (SLE), anti-nuclear antigens (ANA) - RA ## Footnote Can get more than 1 autoimmune condition e.g. autoimmune haemolytic anaemia can be on its own or in combo w/ SLE
273
Primary vs secondary autoimmune pathologies
1o - direct consequence of autoimmune response 2o - consequence of primary pathology (symptoms) e.g. Hashimoto's thyroiditis primary - destruction of thyroid tissue secondary - hypothyroidsim due to loss of thyroid
274
Describe immunological features of autoimmune disease
Auto-Abs depositied in tissues + formal immune complexes Cellular infiltrate (B & T cells) -> autoreactive CD4+ Th cells involved in ~all autoimmune diseases - support B cell stim to make Ab, amplify macrophage responses, target killing via Tc cells, cytokine production Th17 cells also implicated
275
What cells are sufficient for MS transfers in vivo (EAE)?
Th1 & Th17 specific for myelin basic protein. ## Footnote EAE -Experimental Autoimmune Encephalomyelitis
276
What are 3 autoimmune disease mechanisms Abs are involved in?
Damage or destruction - complement & opsonisation Alteration of function - stimulation, inhibition/blockage of function Deposition of immune complexes
277
Auto-Ab mediated damage ## Footnote Autoimmune mechanism
Can be via: - Complement mediated lysis, MAC lyses rbc -> autoimmune haemolytic anaemia - opsonisation of platelets -> autoimmune thrombocytopenia (bleeding)
278
Auto-Ab mediated alteration of function ## Footnote Autoimmune mechanism
Stimulation: auto-Ab agonist activity at TSH receptor - stimulates hormone production -> short-circuits negative feedback regulation mechanism - Graves disease (hyper-thyroidism) Inhibition: auto-Ab antagonist at receptor -> causes internalisation + degradation of endplate - Myasthenia Gravis Blocking: auto-Ab binds intrinsic factor, so it cannot bind + absorb VitB12 -> lack of rbc + tiredness - pernicious anaemia
279
Auto-Ab mediated deposition of immune complexes ## Footnote Autoimmune mechanism
In SLE, complexes deposit in small blood vessel walls -> inflam reaction - Arthritis, 'butterfly rash', vasculitis, glomerulonephritis - Very rare
280
Are genetics associated w/ autoimmunity?
Yes - genetic concordance of related individuals. In autoimmunity concordance in identical twins is larger than non-identical twins BUT <50% -> large environmental role
281
Polymorphisms for autoimmunity ## Footnote More than 1 phenotypic form
- Structural polymorphism: different forms of protein made (coding region of gene) e.g. MHC - Non-structural polymorphism: altered protein activity/protein levels (if in non-coding region promoter/enhancer) Expression of polymorphic alleles -> autoimmunity NOT mutations
282
Relative risk (RR)
Degree an allele of a gene increases susceptibility e.g. HLA-DRB1 gene is strong risk factor for RA Complex multi-gene interactions increase genetic risk
283
Sex related gene as risk factor for autoimmunity
75% autoimmune diseases found in females, arises child-bearing years Ankylosing Spondylitis is one of few diseases more common in males.
284
How does the environment affect autoimmunity?
- vitD, active form may suppress Th17 development - Drugs/metabolites can bind self-Ag + so Ag appears foreign - Toxins cause damage + expose areas for auto-Ab mediated damage - Microbiota/hygiene -> specific commensal (lack of) bacteria - Pathogens can resemble self-antigens + interact w/ risk genes
285
EBNA2 for autoimmunity
EBV nuclear antigen 2 is TF that interacts w/ several genetic risk loci associated w/ lupus - also T1DM, RA & MS NK cells & specific CD8+ T cells associated w/ raised MS risk
286
How are T cells bypassed for autoimmunity?
B cell mitogens can stimulate auto-Ab production in absence of Th cells e.g. bacterial LPS -> plasma cell differentiation. - Not Ag-specific - LPS is acting mitogen - Usually transient
287
How can T cell tolerance be lost (T cell help be gained) for autoimmunity?
Molecular mimicry between pathogens + self-Ag Rheumatic fever - streptococcal cell wall M-antigen stimulates Abs, can also react to heart myosin, joints & kidney.
288
How can drugs,cheicals induce autoimmunity?
Binds self-antigen, modifying it to created T cell epitope that appears foreign, enabling T cells to help autoreactive B cells Normally finite but genetic predisposition can trigger Th cells for self-Ag -> chronic autoimmunity
289
How does chronic autoimmune disease develop?
Via positive feedback from inflammation -> inability to clear antigen. Broadening immune response - epitope spreading (hidden/cryptic epitopes revealed)
290
What is a type 1 hypersensitivity?
Allergy - excessive immune response to harmless antigen, usually specific + restricted. Systemic responses can result in death via anaphylaxis ## Footnote ~20-25% UK population
291
Asthma
Type 1 airway hypersensitivity -> lung epithelium inflammation, smooth muscle contraction, mucus production - IgE mediated, severe bronchiole constriction
292
What 2 processes cause a type 1 allergic reaction?
- Sensitisation of Ag specific IgE - Re-exposure & activation of inflam response
293
How is an Ag specific IgE sensitised?
1. exposure to allergen -> stimulation of Th2 cells, make IL-4 + help produce IgE Ab 2. B cells produce IgE that recognises allergen 3. IgE circulates in blood + associates w/ mast cells in mucosal & connective tissues via FcεR - receptor to constant region of IgE
294
Re-exposure & activation of inflma response in allergic reaction
- Allergen binds IgE on mast cells -> release inflammatory mediators when IgE /FcεR cross-linked by allergen binding - Mast cells release histamine, cytokines, prostaglandins + leukotrienes - Causes vascular permeability, inflammation, muscle contraction, mucus production, immune cell recruitment, swelling, redness, blistering, itching
295
What cells are activated during an allergic reaction?
Mast cells - release granules w/ histamine etc. Eosinophils (dominant allergy ft) also have FceR + can release IL-4 - need IL-5 for their development
296
What effect does mast cell granule release have on GI tract, airways + blood vessels?
GI tract - increased fluid secretion, expulsion of content Airways - decreased diamater + increased mucus production -> congestion + breathlessness, swelling Blood vessels - increased flow + permeability -> increased effector response in tissues (inflam)
297
Anaphylaxis
Systemic response to an allergen -> triggered by large release of histamine. - Dilation of local blood vessels -> drop in blood pressure + consciousness - Difficulty breathing as airways narrow - Oedema or swelling into surrounding tissue ## Footnote Fatal if not dealt with
298
Are food allergies common?
Rarer than other type 1 insensitivities, approx 1/50 children. 46% anaphylaxis deaths '92 ->2018 from peanut/mut allergies - reduced when introduced to infants diets at 4 months
299
Describe type 2 insensitivities
IgG mediated, react to antigens on cells or tissues
300
What are major clinical syndromes of type 2 insensitivities?
Blood transfusion reactions: IgM binds transfused RBC (complement) -> fever, chills, vomiting Haemolytic disease of newborn: Rh- mum + Rh+ partner, baby + mum blood can mix -> sensitisation Prevented w/ anti-Rh Ag antibodies to bind any free foetal RBCs + destroy them before sensitisation. Haemolytic anaemia ## Footnote Can also be drug induced
301
Describe type 3 insensitivity
IgG mediated, against soluble Ag in immune complex w/ Ab. Local - repeated inhalation of antigen e.g. Pigeon Fancier's disease (dried faecal antigens), Farmer's lung (mould spores in hay) Systemic - infection e.g. malaria or in response to serum administration (serum sickness), SLE
302
What is serum sickness?
Systemic type 3 insensitivity. Animals serum stimulates Ab response (2 doses), Ab to serum proteins form complexes -> deposited in blood vessels, can cause fever, rash, arthritis, kidney disorder.
303
Describe type 4 insensitivity ## Footnote Delayed type (DTH)
Contact, allergic reaction after skin contact w/ allergen. Cell mediated - no Ab, instead has Th, Tc + macrophages Takes 24-72hrs -> swelling, blistering + itching e.g. poison ivy, nickel, hapsten responses
304
Describe different methods of allergy testing
Skin prick - lancet introduces allergen to epidermis -> wheal + flare reaction within mins, late phase reaction 4-6 hrs IgE presence using ELISA - if +ve, person atopic (predisposed) to allergy, may be no clinical symptoms Patch tests - allergens applied topically & left for 24-72 hrs to assess type 4 hypersensitivity
305
Does genetics play a role in allergy?
YES - likelihood increases w/ 2 atopic parents, concordance of atopy is 60% in identical twins (env also has role) MHC e.g. HLA-DR5 associated w/ birch pollen, non-MHC-genes (IL-4, FcεR ) epithelial barrier function.
306
Environmental role in allergy
Prevalence of allergy + asthma has increased dramatically over last 50 yrs - less exposure to antigens when children. Contributed by increasing Th2 polarising substances, pollutants, urbanisation, hygiene? -> exposome.
307
Hygiene hypothesis
1989, Strachen Children raised in large families in country less likely to have allergy/autoimmune disease. - Exposure to environmental microbes when young may help prevent hypersensitivities.
308
Old friend's hypothesis
Evolved from hygiene hypothesis. - Reduced microbial diversity in our microbiome due to wester diets + urbanisation - Reduction in worm infections Babies given antibiotics early on -> more likely to develop allergies (altered microbiome). Low asthma prevalence in pops exposed to high microbial environment. Link between eosinophils in asthma due to altered exposure of short chain fatty acids derived from microbiome.
309
Why do we have a Th2 immune response?
Th2 & IgE responses deal with parasitic worms, so evolved to deal with them. Deworming campaign in Kenya - increased eczema in dewormed children (atopic dermatitis) - may play role in allergy mediation
310
Evidence FOR environmental hypotheses for allergy
- correlation of higher incidences of allergy where lower parasite infections - european divide: more allergic diseases in indutrialised countries (Sweden) vs rural countries (estonia) - rural vs urban epidemiological studies
311
Evidence AGAINST environmental hypotheses for allergy
- South/latin america has more asthma than spain/portugal depsite similar culture + poorer living conditions (but high pollution) - worm infections common in parts of USA - asthma increasing in African countries in poverty - industrialised nations not that clean -> pollutants of air + water ## Footnote Also dependent on climate changes -> pollen
312
Allergy treatments in common current use
- Anti-histamines - Steroid anti-inflam drugs - Adrenaline/epinephrine (Epi-Pen) - vasoconstriction blood vessels
313
Low dose allergen therapies
Desensitisation - intra-muscular injection of low dose allergen, monthly 2-5 yrs -> raise IgG rather than IgE
314
mAb allergen therapies
Duplimab blocks cytokine (IL-4) that promotes Th2 differentiation Benralizumab targets IL-5R prevent eosinophil/basophile development + survival
315
Oral tolerance for allergies
Exposure to antigens leads to T cell tolerance, low doses ingested antigen can re-train immune system (high tolerance of gut) e.g. Palforzia (AR101) - peanut derived protein used to redirect immune response
316
List areas of allergic mechanisms being tested for potential treatments
Eosinophilic inflam - anti-IL-5/R, anti-IgE, anti-IL-13, anti-DP2 T1/T17 neutrophilic inflam - anti-IL23, anti-IL17, anti-CXCR2
317
Worm therapy
Worms have evolved to redirect our immune response so could they dampen inappropriate responses Human trials - hookworm infection to treat asthma, investigated for coeliac disease May simply be able to purify parasitic products and use to modulate immune response.
318
Why do T cell developmental stages occur at different regions of the thymus?
Allows varied selection criteria at different checkpoints during tolerance generation. Quality check (DN3) - subcapsualr region Positive selection (DP) - cortex Negative selection (SP) - in medulla (has med ep cells)
319
Main effector functions of Fc binding FcR
Antibody-dependent cellular cytotoxicity (ADCC) Antibody-dependent cellular phagocytosis (ADCP) Complement-dependent cytotoxicity (CDC)
320
FcyR
All bind IgG1 w/ highest affinity. I - resp burst + reactive O2 species, release cytotoxic granules II - eosinophil granule release, B2 -> phagocytosis, both Bs have immunomodulatory tyrosine inhibitory motifs (ITIMs) III - recruits NK cells via intracellular signalling through y or c domain ## Footnote Most common
321
Role of Fc neonatal receptor (FcRn)
IgG transport + recycling to extend of its half life. Binds IgG Abs + serum albumin w/ high affinity at low pH - recycles IgG -> enhance mAb drug efficacy
322
Antibody-Dependent Cellular Cytotoxicity (ADCC)
- Effector cells are all leukocytes which express Fc receptors and contain cytotoxic granules - Three types of Fc gamma receptor are involved: FcγRI, FcγRII + FcγRIIIA - Mechanisms of killing include release of cytotoxic granules containing granzyme and perforin, and generation of reactive oxygen species (ROS) to induce apoptosis of the target cell
323
Antibody-Dependent Cellular Phagocytosis (ADCP)
Clearance of pathogens via pahgocytosis. Ab binds activating FcyRs on phagocytes -> actin cytoskeleton remodelling - forms phagosome -> pro-inflam cytokine/chemokine release
324
What function do fucosylation & aglycosylation provide?
Reduce effector functions of mAb treatments induced by Fc receptor interaction. Aglycosylation - removes N-linked glycan (N297) which normally binds FcyR + activates complement Fucosylation - fucose added -> steric hindrance preventing IgG binding FcyRIIIA, sailic acid addition also reduces binding affinity -> prevents ADCC + CDC
325
Etanercept
Fusion protein to treat RA. Fucosylation. -> highly sialylated IgG mediates immunosuppressie efefcts via DCs
326
What are BiTEs?
Bispecific T cell engagers Made of aglycosylated anti-TAA + anti-CD3 2/ engineered disulphide bridge (stability) Allow simultaneous engagement of: - tumour associated antigens (TAAs) on cancer cells - CD3 on T cells -> activation ## Footnote Could be inactive + actvated in circulation -> slow Ab clearance enhancing clinical benefits