7 autoimmunity 2 Flashcards

1
Q

What happens when developing autoreactive B cells bind self-antigen in the bone marrow?

A

They undergo deletion (death) or inactivation to prevent autoimmunity.

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

What happens when developing autoreactive T cells bind self-antigen in the thymus?

A

They are deleted or inactivated to maintain immune tolerance.

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

What is the role of Notch signalling in T cell development?

A

Notch signalling commits precursors to the T-cell lineage and initiates T-cell receptor gene rearrangement.

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

What happens to immature T cells that recognize self-MHC?

A

They receive survival signals; those strongly recognizing self-antigen are removed.

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

Where do mature T cells migrate?

A

They travel to peripheral lymphoid organs to encounter foreign antigens.

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

What happens when T cells encounter foreign antigens in lymphoid organs?

A

They become activated, proliferate, and eliminate infections.

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

What is central tolerance?

A

A mechanism that deletes or edits autoreactive T and B cells during development in the thymus and bone marrow.

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

What is antigen segregation?

A

A physical barrier preventing self-antigen access to lymphoid organs (e.g., in the thyroid, pancreas).

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

What is peripheral anergy?

A

Inactivation of autoreactive cells due to weak signalling without co-stimulation.

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

How do regulatory T cells contribute to tolerance?

A

They suppress immune responses via cytokines and intercellular signals.

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

What is functional deviation in immune tolerance?

A

It involves regulatory T cell differentiation to limit inflammatory cytokine secretion.

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

What is activation-induced cell death?Apoptosis of autoreactive immune cells in secondary lymphoid tissues.

A

Apoptosis of autoreactive immune cells in secondary lymphoid tissues.

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

What two signals are required for naive T cell activation?

A

TCR/pMHC interaction; 2) CD28/B7 interaction from an activated APC.

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

What two signals are required for T-dependent B cell activation?

A

BCR/antigen interaction; 2) CD40/CD40-L interaction from an activated T cell.

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

What are natural autoantibodies?

A

IgM autoantibodies produced by B1 B cells, usually low-affinity and rarely pathogenic.

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

Why do some autoreactive B2 B cells require CD4+ T cell help?

A

To produce autoantibodies, as many are anergic upon leaving the bone marrow

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

What is the nature of disease-associated autoantibodies?

A

They are often high-affinity IgG with extensive somatic mutations, indicating T cell involvement.

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

What makes some individuals prone to autoimmune diseases?

A

A combination of genetic susceptibility and environmental triggers.

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

What role do genetic factors play in autoimmune disease?

A

They significantly contribute but are not the sole cause (<100% twin concordance).

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

What does polygenic mean in the context of autoimmune disease?

A

Multiple genes contribute to disease susceptibility.

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

Why are many autoimmune diseases more common in females?

A

Possible hormonal effects.

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

What is the association between MHC alleles and autoimmune diseases?

A

Different MHC class II alleles are linked to various autoimmune diseases.

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

What is the significance of HLA-B27 in ankylosing spondylitis (AS)?

A

People with HLA-B27 are 87.4 times more likely to develop AS, but most do not.

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

How do MHC molecules influence autoimmune disease?

A

They determine the peptides presented to T cells, influencing immune responses.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are the major genetic susceptibility loci for Type 1 diabetes (T1D)?

A

HLA-DQ2/DQ8, CTLA-4, and PTPN22

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is the role of AIRE in immune tolerance?

A

It promotes negative selection of self-reactive T cells; mutations cause APECED.

27
Q

What does Fas do in immune regulation?

A

It induces apoptosis of autoreactive cells; defects cause ALPS.

28
Q

What is FoxP3’s role in immune function?

A

It regulates Tregs; mutations cause severe autoimmunity.

29
Q

How does CTLA-4 regulate T cell activity?

A

It competes with CD28 for ligands to inhibit T cell activation

30
Q

How can environmental factors contribute to autoimmunity?

A

Smoking, vitamin D levels, infections, and microbiome composition may play roles.

31
Q

What is molecular mimicry?

A

A mechanism where microbial antigens resemble self-antigens, triggering autoimmunity.

32
Q

Give an example of molecular mimicry.

A

Rheumatic fever: Streptococcal M protein cross-reacts with heart tissue.

33
Q

How do PAMPs contribute to autoimmunity?

A

They activate APCs, increasing the chance of autoreactive T cell activation.

34
Q

How can self-antigens trigger autoimmune responses?

A

Unmethylated CpG DNA (SLE) and ECM fragments (RA) can activate TLRs

35
Q

What is the role of immune privilege in autoimmunity?

A

Damage to immune-privileged sites can expose hidden antigens, triggering autoimmune reactions.

36
Q

How is multiple sclerosis (MS) initiated?

A

An unknown trigger causes inflammation, allowing immune cells to attack CNS myelin.

37
Q

What are post-translational modifications, and how do they contribute to autoimmunity?

A

Alterations in proteins (e.g., citrullination in RA) make them immunogenic.

38
Q

How can smoking increase RA risk?

A

It induces citrullination of proteins, potentially leading to autoimmunity.

39
Q

What is the role of microbes in autoimmunity?

A

They may induce or prevent autoimmune diseases by influencing immune responses.

40
Q

What is the relationship between gut microbes and T1D?

A

Differences in gut microbiota may influence disease development.

41
Q

How does the microbiome influence T1D in mice?

A

Male mice have protective gut microbes that reduce diabetes risk.

42
Q

What are the key treatment approaches for autoimmune diseases?

A

Anti-inflammatory drugs, immunosuppressants, and biologic therapies.

43
Q

Give an example of a cytokine-targeting therapy.

A

TNF-alpha blockade (e.g., infliximab) used in RA.

44
Q

What is an example of a B cell depletion therapy?

A

Anti-CD20 monoclonal antibodies (e.g., rituximab) used in RA and MS.

45
Q

How do inverse agonist peptides work in tolerance induction?

A

They modulate T cell responses to restore immune tolerance.

46
Q

What does GWAS reveal about autoimmune diseases?

A

It identifies genetic variants linked to disease risk, though their functions are often unclear.

47
Q

Why do identical twins not always develop the same autoimmune disease?

A

Environmental and stochastic factors influence disease onset.

48
Q

What is the role of autoreactive CD4+ T cells in autoimmunity?

A

They must escape tolerance mechanisms for disease to develop.

49
Q

What is the two-step process required for autoimmunity?

A

olerance breakdown + environmental trigger.

50
Q

What are key questions in autoimmune disease research?

A

Why do tolerance mechanisms fail?

How do genetics and environment interact?

How can treatments restore immune balance?

51
Q

Describe the full process of T cell development and selection.

A
  1. Bone Marrow: T cell precursors originate here.
  2. Thymus Entry: Precursors commit to the T-cell lineage via Notch signalling.
  3. β-Selection (DN stage): Cells rearrange TCR β-chain; successful rearrangement leads to proliferation.
  4. αβ TCR Formation (DP stage): CD4+CD8+ T cells rearrange their TCR α-chain.
  5. Positive Selection: T cells binding self-MHC receive survival signals.
  6. Negative Selection: Strong self-antigen recognition leads to apoptosis.
  7. Lineage Commitment: Surviving cells become CD4+ (MHC II) or CD8+ (MHC I) T cells.
  8. Exit to Periphery: Naïve T cells migrate to secondary lymphoid organs for activation.
52
Q

Outline the full process of B cell development and central tolerance.

A
  1. Bone Marrow Development: Pro-B cells undergo heavy chain (VDJ) recombination.
  2. Pre-B Cell Stage: Successful μ heavy chain pairing with a surrogate light chain leads to light chain recombination.
  3. Immature B Cell Stage: Express IgM on the surface.
  4. Central Tolerance:
    - Strong self-antigen binding → Receptor editing (light chain rearrangement) or deletion (apoptosis).
    - Weak/no self-antigen binding → B cell survives.
  5. Mature B Cell Stage: Express IgM & IgD, exit to peripheral lymphoid tissues.
53
Q

Explain the process of naïve T cell activation and differentiation.

A
  1. Antigen Recognition: Naïve T cells scan APCs in lymph nodes.
  2. Signal 1: TCR binds peptide-MHC on APC.
  3. Signal 2 (Co-stimulation): CD28 binds B7 (CD80/86) on APC.
  4. Signal 3 (Cytokines): APC cytokines direct T cell differentiation.
  5. Proliferation & Differentiation: T cells undergo clonal expansion and become:
    Th1 (IFN-γ, IL-12) → Activate macrophages.
    Th2 (IL-4, IL-5, IL-13) → Activate B cells.
    Th17 (IL-17, IL-22) → Recruit neutrophils.
    Treg (TGF-β, IL-10) → Suppress immune response.
    CD8+ Cytotoxic T Cells → Kill infected cells.
  6. Effector Function: Activated T cells exit lymph nodes to target infection.
  7. Contraction Phase: Most activated T cells undergo apoptosis, leaving memory cells.
54
Q

Describe the process of B cell activation and antibody production.

A
  1. Antigen Binding: BCR binds antigen in lymphoid follicles.
  2. Internalisation & Presentation: Antigen is processed and presented via MHC II.
  3. T Cell Help (T-dependent Activation):
    CD4+ T cells provide CD40/CD40L interaction and cytokines.
    B cells proliferate in germinal centres.
  4. Somatic Hypermutation & Affinity Maturation:
    In dark zone, B cells mutate their BCR genes.
    High-affinity B cells are selected for survival in the light zone.
  5. Class Switching: B cells switch from IgM to IgG, IgA, or IgE, depending on cytokines.
  6. Plasma Cell & Memory Formation:
    Some B cells become plasma cells (antibody secretion).
    Others become memory B cells for faster future responses.
55
Q

What are the full processes that prevent autoimmunity?

A

central tolerance and peripheral tolerance

56
Q

what is central tolerance?

A

T cells (thymus) → Negative selection (apoptosis) if strongly self-reactive.
B cells (bone marrow) → Receptor editing, deletion, or anergy.

57
Q

what is peripheral tolerance?

A

Anergy: T cells receive signal 1 without co-stimulation and become inactive.
Regulatory T cells (Tregs): Suppress self-reactive T cells via IL-10, TGF-β.
Antigen Segregation: Physical barriers prevent immune system access to certain self-antigens (e.g., eye, brain).
Activation-Induced Cell Death (AICD): Repeated activation triggers Fas/FasL apoptosis in autoreactive cells.

58
Q

Explain how autoimmune diseases develop.

A

Genetic Susceptibility:
Certain HLA alleles increase risk (e.g., HLA-B27 in AS).
AIRE mutations lead to defective negative selection.
CTLA-4 polymorphisms reduce T cell regulation.
Environmental Triggers:
Molecular Mimicry: Pathogens express antigens resembling self-proteins (e.g., rheumatic fever).
Release of Sequestered Antigens: Trauma exposes immune-privileged self-antigens (e.g., sympathetic ophthalmia).
Microbial Activation of APCs: Infections upregulate B7/CD80, leading to bystander activation of autoreactive T cells.
Post-Translational Modifications: Smoking-induced citrullination triggers RA.

59
Q

What are key costimulatory and inhibitory signals in T cell activation?

A

Costimulatory:
CD28/B7 → Essential for full T cell activation
ICOS/ICOS-L → Supports effector T cell function
Coinhibitory:
CTLA-4/B7 → Competes with CD28, dampens activation
PD-1/PD-L1 → Suppresses chronic immune responses

60
Q

What are the main T cell subsets involved in autoimmunity?

A

Th1 → Produces IFN-γ, activates macrophages (e.g., RA, MS)
Th17 → Produces IL-17, recruits neutrophils (e.g., psoriasis, IBD)
Tregs → Suppress autoimmunity via IL-10, TGF-β
Th2 (less relevant) → Associated with allergies, not autoimmunity

61
Q

How are autoreactive B cells controlled in the periphery?

A

Lack of T cell help → Leads to anergy (unresponsiveness)
Fas/FasL-mediated apoptosis → Removes B cells in germinal centres
Regulatory T cells (Tregs) → Suppress autoreactive B cells

62
Q

What is epitope spreading and how does it contribute to autoimmunity?

A

Initial immune attack targets one antigen, but as tissue damage progresses, new self-antigens are exposed.
Example: In SLE, immune responses expand from DNA to histones and ribonucleoproteins.

63
Q

How do cancer immunotherapies cause autoimmune side effects?

A

Anti-CTLA-4 (ipilimumab) and anti-PD-1 (nivolumab) remove immune “brakes,” allowing strong T cell responses.
This can break self-tolerance, leading to colitis, thyroiditis, myocarditis, etc.

64
Q

What is the hygiene hypothesis and how does the microbiome affect autoimmunity?

A

Hygiene Hypothesis → Fewer infections in childhood shift the immune system towards autoimmunity & allergy.
Gut microbiome imbalances are linked to diseases like MS, RA, and type 1 diabetes.
Example: NOD mice (T1D model) show that gut microbes influence diabetes risk.