Autoimmunity (Findlay) Flashcards

1
Q

CD4+ T Cell Activation Process?

A

APC recognizes a pathogen

internalization and antigen processing

peptide presented via MHC class II

Interaction of the TCR complex with the peptide.

A danger signal/ co-stimulation

T cell proliferation/cytokine response

T cell signal to B cells and CD8 T cells to launch an immune response

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

What causes autoimmunity?

A

Autoimmune diseases occur when:

  • The usual control processes are interrupted, allowing lymphocytes to avoid suppression
  • When there is an ‘alteration’ in cells/tissue so that it is no longer recognised as “self” and is thus attacked.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is “Tolerance”?

A

Tolerance is the prevention of an immune response against a particular antigen

For instance, the immune system is generally tolerant of self-antigens, so it does not usually attack the body’s own cells, tissues, and organs

The immune system also usually ignores (is tolerant to) food antigens

When this “tolerance” to self breaks down it can give
rise to autoimmunity

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

What are the two types of tolerance?

A

Central tolerance
- Thymic selection and regulation

Peripheral tolerance
- Post-thymic selection and regulation

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

Steps of T cell selection in the thymus?

A

Must Express TCR (T Cell Receptor):
- T cells must express a functional TCR to progress in their development
- The TCR is essential for recognising peptide antigens presented by Major Histocompatibility Complex (MHC) molecules on other cells

Positive Selection:
- This process ensures that T cells with TCRs capable of recognising self-MHC molecules (either Class I or Class II) are selected to survive
- T cells that cannot recognise MHC molecules fail to receive necessary survival signals and die by apoptosis (programmed cell death)

Negative Selection:
- Negative selection involves the elimination of T cells that bind too strongly to self-antigens presented by MHC
- Also eliminates T cells that recognise self-peptide in self-MHC
- This process prevents autoimmune responses by deleting T cells that could potentially recognise and attack the body’s own tissues.

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

How does positive selection work?

A

Thymic T cells live 3-4 days unless rescued by TCR engagement

Only small % of all generated TCRs will ever be able to functionally recognise MHC molecules - these are chosen to continue

Mouse thymus produces 50 million DP thymocytes per
day - 10% survive positive selection

Want to find cells which recognise self-MHC

Self-MHC presented by cortical thymic epithelial cells
(cTECs)

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

How are T cells activated?

A

TCR recognises peptides in the cleft of MHC molecules on surface of APCs and then signals through associated CD3 complex proteins

Peptide + MHC class I:
- CD8+ T Cells

Peptide + MHC class II:
- CD4+ T Cells

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

What does negative selection do?

A

Following positive selection, we have T cells which we know can signal through CD3 and recognise MHC. We need to have a way to reject cells which:

  • Recognise self MHC strongly
  • Recognise self peptide

As these cells are likely to induce autoimmunity in the periphery

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

What is AIRE?

A

Autoimmune Regulator (AIRE)

Promotes the expression of diverse self-antigens in the thymus

What this means is that in a normal human thymus, on epithelial cells, every single type of antigen that you might ever have inside your body, can be expressed on the MHC

Essential for the negative selection of T cells

Helps eliminate T cells that are reactive to the body’s own tissues, reducing the risk of autoimmune diseases

Maintains immune self-tolerance and prevents the immune system from attacking the body’s own cells

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

What is APS1/APECED?

A

It is an autoimmune disease

“Autoimmune Polyendocrine Syndrome type 1” /
“Autoimmune PolyEndocrinopathy-Candidiasis-Ectodermal Dystrophy”

Caused by mutations in the AIRE gene

Patients present in childhood or teenage

Requires person to have 2/3 of the following:
- Candidiasis
- Addison’s disease
- Autoimmune hypoparathyroidism

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

Antibodies structure

A

Y-shaped molecule composed of four polypeptide chains.

Two identical heavy chains and two identical light chains.

Variable (V) Regions: Located at the tips of the Y, specific to each antibody, determine antigen binding specificity

Constant (C) Regions: Part of the chains that determine the antibody’s class and its role in the immune response

Disulfide bonds link the chains together and stabilise the antibody structure

Contains a flexible hinge region which allows flexibility and variability in positioning when binding to antigens

Five classes:
- IgG, IgM, IgA, IgE, IgD, classified based on differences in the constant region of the heavy chains

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

What is B cell central tolerance?

A

Occurs in the bone marrow, where B cells mature

Involves negative selection of B cells that bind strongly to self-antigens

2 Processes:
- Receptor Editing: B cells that recognise self-antigens may rearrange their receptor genes to alter specificity.

  • Clonal Deletion: B cells with receptors that still recognise self-antigens are eliminated

Binding of self reactive IgM to self antigen in the bone marrow leads to death of the cell. Estimated to be approx. 75% of B cells killed in this way

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

What happens when a B cell encounters a multivalent self molecule during central tolerance?

A

A multivalent self molecule has multiple identical epitopes.

Strong cross-linking of the B cell receptor (BCR).

Typically leads to strong signalling for negative selection, such as clonal deletion or receptor editing, to prevent autoimmunity.

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

What is the effect of encountering a soluble self molecule on B cell central tolerance?

A

Soluble self molecules are unattached and free-floating in the bone marrow environment.

May bind to BCR without cross-linking.

Often results in receptor editing to change specificity or, if the interaction is of high affinity, clonal deletion.

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

How does a B cell respond to a low-affinity non-cross-linking self molecule during central tolerance?

A

These molecules bind weakly and do not cause significant receptor clustering.

May lead to B cell anergy, where the cell becomes functionally inactive but is not deleted, or the B cell may escape deletion due to insufficient signalling for negative selection.

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

What occurs if a B cell does not react with any self molecule during central tolerance?

A

B cell receptors do not recognise or bind to any self antigens.

The B cell is considered non-self-reactive and successfully matures, exiting the bone marrow to participate in the peripheral immune response, enhancing immune diversity and readiness.

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

What is the major control mechanism in the periphery?

A

T regulatory cells (Tregs)

Natural Tregs are called nTregs
Thymus derived Tregs are called tTregs

These are a bridge between central tolerance and peripheral tolerance

They are cells that are generated in the thymus but which act in the periphery

Tregs can catch the damaging self-reactive T and B cells which escape negative selection

17
Q

What is CD25+?

A

CD25 is notably expressed on regulatory T cells (Tregs) and is used as a marker to identify these cells

Tregs are essential for maintaining immune tolerance and preventing autoimmune responses.

When CD25+ cells are knocked out of mice, 100% get autoimmune disease

18
Q

What is foxp3?

A

FOXP3 is a transcription factor that is crucial for the development and function of Tregs

It regulates the expression of genes that are necessary for the suppressive activity of Tregs`

The expression of FOXP3 essentially defines Tregs and is required for their development and suppressive functions

It acts to modulate the immune response and maintain self-tolerance

When FOXP3 is knocked out of mice models, their organs filled up with T cells and they die

19
Q

How do Tregs work?

A

They make suppressive inhibitory cytokines

They can directly kill cells by releasing granzyme (cytolysis)

They can produce adenosine to cause metabolic disruption

They can prevent dendritic cells from maturing

20
Q

What happens with a FOXP3 mutation?

A

A disease called IPEX

“Immune dysregulation polyendocrinopathy X-linked”

Mutation in foxp3 causes no natural Treg cells

Suffer from:
- Severe allergic inflammation
- Polyendocrinopathy
- Enlargement of lymphoid organs
- Diabetes
- Anaemia
- Early death

21
Q

What is the T cell effector functions?

A

Main targets:
- Clears pathogens
- Kills infected cells
- Recruits and activates other immune cells to clear up damage

But can also result in:
- Attacks on self antigen
- Releases cytokines which damage host tissue
- Occurs in inappropriate sites
- Over reacts to innocuous challenge

So needs to be kept in careful balanceW

22
Q

What are the 5 main mechanisms to keep T cells in control?

A
  1. Ignorance
  2. Anergy
  3. Negative co-stimulation
  4. Activation induced cell death
  5. T regulatory cells
23
Q

What is ignorance?

A

Ignorance (immune privilege)

Immune privileged sites which do not spark immune responses

Grafts in these sites are not rejected

Have barriers that exclude naïve T cells

Produce anti-inflammatory cytokines eg TGFbeta

Lymphocytes which do enter are killed (Fas)
- E.g. brain, eye, testis, foetus

24
Q

What is sympathetic autoimmune opthalmia?

A

Eye usually experiences ignorance, with no T cells present in the eyes, so any antibodies in the eyes are never detected and immune response never occurs

Sympathetic autoimmune opthalmia is trauma to one eye resulting in the release of sequestered intraocular protein antigens

Released intraocular antigen is carried to lymph nodes and activates T cells

Effector T cells return via bloodstream and encounter antigen in both eyes

25
Q

What is induction of anergy?

A

Induction of anergy refers to the process where immune cells, particularly T cells, become non-responsive to their specific antigen, preventing an immune response even in the presence of that antigen

Some autoreactive T cells escape the thymus e.g. Multiple
Sclerosis

The requirement for co-stimulation in the T cell activator process in the periphery means T cells are not activated by continual self antigen presentation by DC

This requirement is also harnessed to suppress self-reactive T cells through induction of ANERGY

Anergy defined as ‘ failure to proliferate or produce IL-2
following presentation of cognate antigen’

26
Q

Anergy in T cell mechanism

A

Naive T cells which recognise self antigen in the absence of co-stimulation enter state of anergy because there is no inflammation driving DC maturation, so no need to respond

27
Q

What is negative co-stimulation?

A

It is the switching off of T cell activation via negative co-stimulation

Some co-stimulatory molecules are negative rather than positive signals

PD-1 ,CTLA-4 are the main ones

28
Q

What is CTLA-4?

A

CTLA4 is a major negative regulator

It is initially intracellular but moves to cell surface after TC signalling

It binds to the same ligands as CD28, but with a higher affinity for CD80 / CD86 than CD28

It binds all the CD80/86 so prevents CD28 signalling

These negative co-stimulators are the target of cancer therapies - where we want lots of very activated anti-tumour T cells

29
Q

What is activation induced cell death? (AICD)

A

After the effector T cells have activated other cells or killed
their targets, they must be removed

This is done by Fas / Fas ligand interaction - called activation induced cell death

T cells express Fas as they are activated. When T cells bind other Fas presenting cells (can be T cells), Fas is trimerized and activates the apoptosis pathway

T cells can also express FasL and kill each other

30
Q

What is Autoimmune Lymphoproliferative Syndrome ALPS?

A

It is a autoimmune disease caused by mutations of Fas or downstream Fas pathway

Leads to build up in lymph

Can lead to lymphoma

31
Q

What mechanisms are used by infections to induce autoimmune responses in the host?

A
  • Molecular mimicry
  • Epitope spreading
  • Bystander activation
  • Cryptic antigens
32
Q

What is molecular mimicry?

A

Molecular mimicry is where the antigens of a pathogen resemble the body’s own proteins, causing the immune system to attack its own tissues, triggering autoimmune diseases

E.g.
- Streptococcus pyogenes infection can trigger cardiac pathology

  • T cell clones from heart lesions can recognise
    BOTH heart-specific proteins and streptococcus M protein
33
Q

What is bystander activation

A

Bystander activation is a mechanism where immune cells are activated indirectly due to the inflammatory environment created by an immune response

This can lead to the activation of immune cells that have not encountered their specific antigen

Some of these may be auto-reactive T cells

Inflammation also temporarily suppresses Treg

34
Q

What is hashimoto thyroiditis?

A

Is is the first disease to be recognised as an autoimmune disease

Autoreactive antibodies and T cells that attack the thyroid:
- Inflammation
- Enlargement of thyroid
- Gradual destruction of follicles in thyroid

Blocking antibodies
- Stimulate the thyroid-stimulating hormone (TSH) receptor,
- Thyroid peroxidase and or thyroglobulin

Resulting in inadequate iodine uptake and thyroid hormone production and secretion

Most commonly affects middle age females

Genetics : HLA-DR5, CTLA4

Symptoms:
- Weight gain
- Depression
- Mania
- Sensitivity to cold
- Fatigue
- Panic attacks
- Tachycardia

Hashimoto’s thyroiditis is often misdiagnosed as depression

Testing for-thyroid antibodies can resolve any diagnostic difficulty.

35
Q

What is graves disease?

A

Hyperthyroidism

Graves’ disease is an autoimmune disease characterised by the overproduction of thyroid hormones due to the immune system producing antibodies that stimulate the thyroid gland

Ordinarily, TSH is secreted by the pituitary gland that binds TSH receptors and stimulates the thyroid to secrete thyroid hormone, which acts on the pituitary gland and shuts it down

With graves disease, the pituitary gland is still shut down by the secreted thyroid hormone, however, the antibodies continue to stimulate the thyroid, endlessly producing thyroid hormone

Symptoms include:
- Enlarged thyroid (goiter)
- Bulging eyes
- Increased heart rate
- Weight loss

Treatment often involves medications to reduce thyroid hormone production, radioactive iodine, or surgery.

36
Q

What is systemic lupus erythematosus?

A

Lupus is a systemic autoimmune disease that attacks many tissues.

More common in women than men (9:1).

Symptoms include:
- Fever, arthritis
- Skin rash
- Pleurisy
- Kidney dysfunction

Caused by auto-antibodies to DNA, histones, RBCs

Leads to formation of immune complex and complement activation leading to vasculitis.

37
Q

What are some treatments for autoimmune diseases?

A

Steroids

Symptomatic treatment e.g. insulin

Induce anergy - block T cell co-stimulation

Shut off T cell activation - PD1, CTLA4

Suppressive cytokines

Infuse Treg

38
Q

How do we treat Crohns disease

A

Corticosteroids are used as short-term treatments for disease flares.

They act quickly to reduce inflammation by inducing lymphocyte death and reducing T cell activation and cytokine production

BUT they suppress the entire immune system and have significant side effects

So the ‘biologic’ class of therapy has now been developed

Anti-TNF (infliximab and newer versions) was a game-changer when it was developed

This induces and maintains remission as it prevents release of TNF, the key pro-inflammatory cytokine

BUT this blocks all TNF

Patients are at risk of infections - some severe, including an increased risk in Tuberculosis susceptibility

The Holy Grail is finding a population of pathogenic cells with a marker that can be specifically blocked

39
Q
A