Immunology Flashcards

1
Q

What does immunological tolerance do?

A

Serves to protect us from self-reactive lymphocytes.

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

What tissues comprise central tolerance?

A

Thymus - T cells

Bone marrow - B cells

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

What tissues comprise peripheral tolerance?

A

Secondary lymphoid organs

Peripheral tissue

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

What are the 4 mechanisms used to induce tolerance to self-reactive lymphocytes?

A

To delete (eliminate the problem)

To anergise (switch off the problem)

To ignore (ignore the trigger)

To regulate (contain the problem)

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

What mechanisms are used for central B cell tolerance?

A

Deletion and anergy

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

What mechanisms are used for peripheral B cell tolerance?

A

Ignorance/anergy/death - lack of co-stimulation/T cell help

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

Which is more efficient - T or B cell tolerance?

A

T cell tolerance is more efficient

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

What happens in central B cell tolerance when there’s a low-affinity non-cross-linking self molecule?

A

There’s binding to one arm, but there’s no cross-linking, meaning there’s not enough affinity to trigger the immature B cell. The cell can still mature because it has the antigen receptors on its surface, but because of its low affinity, it is clonally ignorant.

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

What happens to an immature B cell in central tolerance when it encounters a multivalent self molecule?

A

There is extensive cross-linking and rather than turning off the cell, the cell death program is initiated in the immature B cell and it apoptoses.

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

What happens to an immature B cell in central tolerance when it encounters a soluble self molecule, e.g. a serum protein?

A

B cell is triggered, but because it is immature, it migrates to the periphery, resulting in an anergic B cell.

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

In peripheral B cell tolerance, which 2 signals are needed for a mature B cell to respond and survive?

A
  1. Signals via the surface Ig-Ag interaction
  2. T cell help – CD40L, and some cytokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How long is B cell life span in the absence of T cell help?

A

Short

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

When does peripheral B cell tolerance also occur?

A

Post-somatic hypermutation

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

What will happen when a naïve B cell sees an antigen?

A

Naïve B cell can see the antigen

Activation of the CD4 compartment simultaneously, providing the ligand.

Initial response will be crosslinking and a commitment to differentiating into a plasma cell

Because the signals from CD4 aren’t there, it will be short lived with a low affinity antibody and won’t be isotype switched in the domain

B cell dies

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

What happens when a naive B cell sees an antigen as well as CD4?

A

Cells transition into germinal centre of lymph node

Isotype switching

High affinity antibodies from plasma cells or generation of memory

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

What does peripheral B cell tolerance rely on?

A

T cell tolerance working

If a CD4 T cell is not self-reactive then there’s no way a self-reactive B cell is going to be able to get going very well.

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

In the absence of CD4 T cell help, will there be production of antibody from a naive B cell?

A

Yes, but it’s transient

The antigen is low affinity and because the plasma cells are terminally differentiated, it dies off.

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

Why are self-antigens often seen after infection?

A

There is release of self-antigens from damaged cells and it may trigger self-reactive B cells in the periphery.

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

Why won’t self antigens released from damaged cells after infection trigger self-reactive B cells in the periphery?

A

Because there’s no CD4 help that is specific for the self-antigens, there’s no cognate help and therefore no robust response in the domain.

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

How is antigen recognition different between B and T cells?

A

B cells see whole proteins (or components of whole proteins/pathogens)

T cells see peptide fragments that are processed and presented in the context of MHC molecules

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

Why are T cells technically self-reactive?

A

Because they have to see a self antigen (the MHC complex)

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

Where do T cells develop?

A

In the thymus

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

How do T cells develop in the thymus?

A

Early T-lineage precursor receives signals and progresses through different stages to DN thymocyte.

DN thymocytes are commited to T cell lineage but don’t express co-receptors like CD8.

Beta chain is rearranged (checkpoint):

If functional, commited to double positive stage (alpha chain of the alpha-beta heterodimer (the receptor) is expressed at cell surface).

If they see MHC I they will commit to CD8, if they see MHC II they will commit to MHC II.

This is called positive selection.

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

What is positive selection of thymocytes (T cell precursors)?

A

Double positive (DP) thymocytes undergo 2 selection processes following the expression of a TCR

Thymocytes that express TCRs capable of recognising self-MHC are selected to survive.

Only thymocytes that express TCRs capable of recognising self-MHC are selected to survive.

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

What is negative selection of thymocytes (T cell precursors)?

A

Removal of immature lymphocytes that have strong reactivity to self-peptide.

This is the major mechanism by which T cells are deleted if they react too well to self MHC and self-peptides.

Removal of immature lymphocytes that have strong (e.g. too good) reactivity to self peptide.

26
Q

What is T cell selection dependent on?

A

Receptor affinity for self pMHC (Goldilocks Theory)

If there’s not enough interaction (not positive selection) or if the TCR sees self and MHC too well, T cells will die. There’s a sweet spot, where positive selection and commitment to a mature T cell occurs.

27
Q

What is AIRE?

A

Autoimmune regulator of expression.

Some tissue specific antigens are expressed in thymic medullary epithelial cells under control of the AIRE (autoimmune regulator of expression) transcription factor

AIRE randomly distributes itself wherever DNA is accessible or open and can turn on gene expression non-specifically.

28
Q

What does AIRE do?

A

AIRE randomly distributes itself wherever DNA is accessible or open and can turn on gene expression non-specifically. Two things happen here:

Gene expression that is not normally associated with the thymus, e.g. pancreatic tissue, in the medullary epithelial cells. As a consequence, there is processing of the proteins and presentation by MHC class I and if an immature thymocytes sees it, it will be deleted.

Because there’s a massive deregulation of transcription in these cells, death will often be induced. Resident dendritic cells will then pick up the dead cells/debris and present them to an auto-reactive T cell and then there will be deletion.

29
Q

What happens if there’s a defect in AIRE?

A

A defect in AIRE is a defect in the ability to present antigens that would be used to delete T cells, which is why carriers of the mutation present with multisystem/multiorgan autoimmune disease.

30
Q

What does central T cell tolerance involve?

A

Immature/developing lymphocytes and occurs in primary lymphoid organs:

Deletion

Selection of Tregs

31
Q

What does peripheral T cell tolerance involve?

A

Mature lymphocytes and occurs in secondary lymphoid organs and peripheral tissues:

Deletion

Anergy

Ignorance

Regulation

32
Q

What is the schamatic for naive lymphocyte circulation in ignorance?

A

Naive lymphocytes enter lymh nodes from blood

Antigens from sites of infection reach lymph nodes via lymphatics

Lymphocytes and lymph return to blood via the thoracic duct.

33
Q

What is the two signal theory for activation of naive T cells in anergy?

A

In the periphery, 2 main signals are needed to induce a mature T cell response:

Stimulation via the TCR on the T cell

Co-stimulation via CD28 and CD80 on the dendritic cell.

Without co-stimulation, T cell becomes anergic

34
Q

What do Treg cells express?

A

High levels of IL-2 receptor alpha

TGF-beta

IL-10

Expression associated with FOX-P3 transcription

35
Q

What is a key identifier of Treg cells?

A

FOX-P3 (transcription factor)

36
Q

What can Tregs do?

A

Suppress all manner of helper T cell and CD8 cell responses.

It’s not just in terms of tolerance that they’re important but also to help attenuate inflammatory responses against viral infections.

37
Q

What are the most prominent suppressors of immune suppression?

A

Tregs

38
Q

What are the 2 different types of Tregs?

A

nTregs (natural) – derived from the thymus during T cell development

iTregs (induced) – derived following activation of naïve CD4 T cells in the presence of TGF-beta

39
Q

What do iTregs do?

A

Secrete immunosuppressive cytokines – IL-10 and TGF-beta

Express CTLA4 and inhibit co-stimulation

Release molecules that create a suppressive environment

40
Q

How does CTLA4 work?

A

CTLA4 also binds B7, but it’s also a competitor for CD28 in that it has higher affinity for the costimulatory molecules. As a consequence, if there are high levels or a high concentration of CTLA4 on the cell surface, it can start to inhibit the ability of naïve T cells to sustain their activation.

41
Q

What is autoimmunity?

A

Loss of the tolerance mechanism

42
Q

What happens in loss of tolerance?

A

Immature developing lymphocytes in primary lymphoid tissue that react to self antigens are removed (clonal selection)

Failure to remove the auto-reactive lymphocyte leads to generation of loss of tolerance.

43
Q

Which 3 key components are needed in the pathogenesis of autoimmune disease?

A

Genetic susceptibility: especially for T cell autoimmune diseases, like diabetes or MS.

Environmental: e.g. stress or malnutrition

**Loss of self-tolerance: **Can be induced by either of the two above

44
Q

Why are circulating autoreactive lymphocytes not always activated?

A

Antigen is not available

Absence of signal 2 - To get immune response need danger signal to activate DCs, supply co-stimulation - might get T cell help from somewhere else

45
Q

What is the difference between autoimmune response and autoimmune disease?

A

Continued damage promotes lymphocyte activation. There’s inflammation, so there’s secondary signals and there are going to be self-antigens that might otherwise be sequestered.

See image

46
Q

Do autoimmune responses always result in autoimmune diseases?

A

No

47
Q

Do all autoimmune diseases involve autoimmune responses?

A

Yes

48
Q

What does autoimmunity result from?

A

Chronic (ongoing) autoimmune responses with ongoing tissue damage.

49
Q

What are the 2 classifications of autoimmune diseases?

A

Organ-specific and systemic

50
Q

What is organ-specific autoimmune disease?

A

Confined to particular organs or cell types and the antigens recognized are organ specific.

E.g. thyroid, ovarian, islet cells, gastric parietal cell (pernicious anaemia); neurological: MS, myasthenia gravis

51
Q

What is systemic autoimmune disease?

A

Multiple tissues of the body are targeted and the antigens recognised are more ubiquitous.

E.g. rheumatoid arthritis, systemic lupus erythematosus (SLE)

52
Q

What are the normal effector mechanisms in response to self antigens?

A

B cells – production of auto-antibodies:

Type II (including ligand mediated reactions), immune complex deposition (Type III)

**CD4 and CD8 T cells: **

DTH responses (TH1 activation of macrophages, cytokine production, pro-inflammatory mediator release)

CTL killing of stromal cells

Provision of B cell help

Macrophages

Nitric oxide, proteases, oxidative radicals

53
Q

What can loss of central self tolerance be due to?

A

Autoimmune polyendocrinopathy-candiadiasis ectodermal dystrophy

Defect in the AIRE gene

Decreased central tolerance

Multi-system autoimmunity

54
Q

What can loss of peripheral tolerance be due to?

A

Immunodysregulation, polyendocrinopathy, and enteropathy, X-linked

Defect in the Foxp3 gene

Loss of Tregs and peripheral tolerance mechanism

Multi-system autoimmunity

55
Q

What are some diseases resulting from B cell mediated autoimmunity?

A

Grave’s disease (stimulating Ab)

Myasthenia Gravis (inhibitory Ab)

SLE (immune complex deposition)

56
Q

What are some diseases resulting from T cell mediated autoimmunity?

A

Insulin dependent diabetes mellitus (IDDM)

Multiple sclerosis

57
Q

What happens in Type 1 insulin dependent diabetes mellitus (IDDM) Juvenile onset diabetes?

A

Organ specific, T cell mediated (both CD4 and CD8)

Autoimmune destruction of pancreatic β-cells (insulin producing)

Characterised by infiltration of lymphocytes, weak autoantibody response, T cell reactivity to islet proteins

Gradual loss of insulin secretion, and resultant insulin dependence

Occurs more frequently in people with HLA DR3-DQ2 and DR4-DQ8

58
Q

What happens in multiple sclerosis?

A

Polygenic degenerative disorder of the CNS that results in episodes of paralysis during formative disease, with chronic paralysis in the latter stages

CD4 T cells specific for myelin antigens promote an inflammatory response and degrade the myelin sheaths covering nerve axons

Th1 and Th17 responses are detrimental

Th2 responses associated with remission

Dysregulation of Tregs has been associated with MS

HLA-DR15 and HLA-DQ6 associated with disease

59
Q

How do autoimmune diseases begin?

A

Circulating B cell binds self antigens released from injured cells

B cell is activated by a T cell specific for self peptide

B cells differentiate into plasma cells, secreting large amounts of self-antigen specific antibody

At sites of injury, self-antigen specific antibody initiates an inflammatory response, causing more injury

More B cells bind self antigens, amplifying the cycle of tissue damage

60
Q

What is the role of infection in initiation of autoimmune disease?

A

Infection with microorganism

Get exposure of self antigens due to dying cells

Infection can activate DCs (e.g. TLR signalling), providing co-stimulation

61
Q

What is the role of autoreactive lymphocyte activation in initiation of autoimmune disease?

A

Molecular mimicry

Antigens from pathogen are similar in shape to autoantigens

Able to cross react with autoreactive T cells/B cells