Adaptive Development (B and T Cell Receptor Generation, Tolerance) Flashcards

1
Q

How does endocytosis of an antigen and subsequent presentation allows potential help from T cell which recognise different epitopes?

A

a B cell receptor recognises a peptide sequence in an antigen (6aa long)

help for the B cell can come from any T cell that is specific for the presented peptides, not just the one the B cell initially recognises

important as T cell antigen may be in a deeper part of the protein (out of Ab reach)

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

Why are some regions of the antigen binding site of BCRs non variable?

A

variation is focussed on 3 short segments in the V region w hypervariable aa sequences
- known as complementarity determining regions (CDRs)

CDRS alternate w 4 framework regions (FRs) that provide Ig w structure
– if they were more variable the protein may not fold properly and become non functional

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

What is genomic rearrangement?

A
  1. genomic rearrangement
    – V, D, and J selection determines B cell receptor specificity
    – large number of genes = massive diversity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the significance of B cells possessing a large set of variable regions?

A

– determined by junctional diversity and somatic mutations
– only one generated per B cell
– all Ig from a single B cell have the same specificity

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

What is affinity maturation?

A
  • can increase the binding strength of Abs via somatic mutations in the hyper variable regions
  • improved affinity = better Abs
  • somatic mutations can also create lower affinity Abs, but these will quickly be outcompeted
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is class switching?

A

allows B cells to irreversibly alter the effector function of Abs they produce

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

What is the genetic organisation of light chains in mice?

A

in mice there are two light chains (kappa (k) and lambda)
- each contains V, J, and C

k is used first as it can give rise to highest diversity as it is more complex
- any V gene can join to any J
- 95% of mouse Abs have a k light chain (70% in humans)
- ~350 V, 5 J,

lambda is used as a last resort and cannot generate as much diversity
- gene is split into sections. not any V can join any J, just ones in their section
- 3 V, 4 J

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

Outline lambda chain rearrangement.

A

the joint between the V and J region are always located within CDR3
- tru for k and heavy chains too
- helps further diversity found in Ig binding domains

lowest level of variation (accounts for 5% of Ig in mice)

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

Outline kappa chain rearrangement.

A

If V joins to J1 then J2-5 remain in the intron

If V joins to J5, then J1-4 are excised out from the chromosome and are lost

in this instance, the rearrangement is non-productive (eg the VJ gene is out of frame) then cell will rearrange on other chromosome
- if this also fails, cell will then try on lambda chain

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

What is the genetic organisation of the heavy chain?

A

larger locus and contains additional fragments

heavy chains have a diversity segment (D) which encodes aa within CDR3
- 10-14 segments in mice
- further expands range of epitopes

heavy chain also has multiple constant (C) genes which determine the class of Ab produced
- IgM and IgD in naive B cells (alternative splicing)
- class switching post activation

junctions (//) between regions sites for insertion of random aa

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

What is the order of recombination?

A

heavy chain recombines first
– J combines to D first then cut out remaining Ds to joining on of V
– can only happen once per chromosome as all D genes are cut out

cells will then rearrange kappa light chain
– if unproductive, a rescue attempt can happen if there is a 5’ V and a 3’ J left

lambda is used as last resort
- rescue attempt can be made if there is a 5’ V and a 3’ J left (human
- unlikely for mouse due to structure of genes (sections)

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

Taking into account other additions/ alterations which can affect diversity, there are possibly 10^10/10^11, explain why the body would not actually produce this many different BCRs.

A

a good portion of these BCRs would be unproductive, unreactive, or improperly folded

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

What are the mechanisms of recombination?

A

every V, D, and J genes has conserved flanking sequences
- recombination signal sequences (RSSs)
- found at 5’ and 3’ ends of the genes

highly conserved regions, eg

k structure:
V – (12 bp) – // – (23bp) – J

lambda structure:
V – (23 bp) – // – (12bp) – J

H chain structure:
V – (23 bp) – // – (12bp) – D – (12bp) – // – (23 bp) – J

sequences are palindromic
12 p 1 turn of DNA helix
restriction enzymes target these regions

recombination occurs between a 23 RSS and a 12 RSS
- means each gene is joined to correct gene

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

What is the function of RAG1/2?

A

RAG1 binds to both RSS and histone H3 (part of DNA structure

RAG2 guided by RAG1 and is enzyme w DNA cleavage activity

activity of RAG1/2 form a loop structure and bring the parts to be joined into close proximity

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

Outline the role of RAG1/2, and Ku70:80 in VDJ recombination step by step.

A

1, RAG 1/2 recognise conserved heptamer/nonamer sequences of RSSs

  1. RAG complex at one site (eg D gene) bind to the RAG complex from a different site (eg J gene)
    – this forms a loop of DNA
  2. RAG2 cleaves DNA and forms hairpin loops
  3. DNA repair enzymes Ku70:Ku80 bind DNA at ends and stabilises strand break complexes

5a. signal joint is ligated together to form a loop, which dilutes out as cell divides

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

Outline the role of Artemis and TdT in VDJ recombination.

A

5b. DNA protein kinase and Artemins binds to closed loops of DNA, covering ends of the two loops being recombined
- this cleaves the hairpin loops, introducing P bases, creating raggedy ends
- TdT adds random, non-templated nucleotides, including non-complementary bases
- complementary bases pair before mismatches are repaired
- DNA ligase and XRCC4 edit ends through base excision and repair to fully bind and ligate DNA back together

new section of DNA will always be different even if genes are the same

Artemis and TdT do not act on signal joints, only coding joints

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

What happens if TdT adds too many nucleotides?

A

region will be too long, creating a frameshift in CDR3
– meaning it will be non-functional

generation of greater diversity at a cost

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

Where are promoters and enhancers of Ig genes located?

A

every V genes has a promoter of 25bp at 5’ end that is shared between H and L chains
- results in equal transcription of both

an enhancer is needed to initiate transcription of the gene
- they are only close enough after VDJ recombination events have taken place

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

If recombination is successful on the first chromosome, what happens to the second one?

A

allelic exclusion
- rearrangement is suppressed

ensures B and T cells have a single specificity
- dual is a problem, as cell could have one receptor that recognises non-self and the other recognises self
- if it were to be activated, would cause major autoimmune reaction

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

How is allelic exclusion carried out?

A
  1. initiation
    - allele asynchrony
    • one allele opens up first, the other has mechanisms to keep it closed
  2. feedback
    - after first rearrangement, will recombine w section of protein which creates pre B or pre T receptor = surrogate light/alpha chains that join onto first recombination which allows it to be expressed on surface
    - if it is functional (binds to heavy chain, stable) it will send signals to halt rearrangement of second allele
  3. maintenance
    - second allele is silenced
    - cant be opened up so RAG cant enter
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What is the structure of a TCR?

A
  • no alternative constant regions
  • never secreted
  • heterodimeric joined by a disulphide bond
  • short cytoplasmic tail
  • antigen binding site made up of both subunits
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What does VDJ recombination look like in T cells?

A

the mechanisms that control the recombination events are the same between B and T cells

a chain contains V and J
b chain contains V, D, and J

V has 23
J has 12
D has both

this means that D region can potentially be skipped
- higher diversity

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

What is different between rescue events between T and B cells?

A

rescue can happen on all T cell alleles, but in B cells this will only occur on the light chain

T cell a chain has more options than light chains for rescue of rearrangement events, due to D being able to be skipped on beta
and T cells have two sets of clusters

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

Where is the delta locus located?

A

located within the alpha locus, with the V regions interspersed

some of the V segments are shared between a and d

  • rearrangement of the a chain results in deletion of the d locus from the chromosome

At the DN2/DN3 stages, TCRγ, TCRδ, and TCRβ gene loci start undergoing somatic recombination
Rearrangement of the TCRγ and TCRδ loci occurs simultaneously.
Rearrangement of the TCRβ locus is independent of the γδ loci.

If both TCRγ and TCRδ loci successfully rearrange and produce a functional γδ TCR before the TCRβ locus, the thymocyte expresses the γδ TCR on its surface.
These cells bypass the pre-TCR checkpoint and commit to the γδ lineage.

If a functional TCRβ chain rearranges first, it pairs with the pre-Tα chain to form the pre-TCR complex

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

What are the different outcome of alternative recombination in the delta locus?

A

V-J
V-D-J
V-D-D-J

joining of V-J directly is often limited due to other control mechanisms

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

What is somatic hypermutation?

A

occurs mainly in the CDR regions after clonal expansion, and can happen again after reactivation (secondary/tertiary immune response

mutation rate in variable regions is 10_3 bp/generation (10_8 in other genes)

selection of higher affinity clones for selective expansion
– lower affinity clones are outcompeted and die

enhanced diversity by 1-2 orders of magnitude

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is the mechanism of hypermutation?

A

involved are RNA pol II and activation-induced cytosine deaminase (AID)

AID is responsible for deamination of bases in DNA
- C residue is replaced w U
– doesnt belong in DNA
– must be repaired

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is the cost of hypermutation?

A

many cells will mutate to non-functional Igs, or will lower affinity for receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Why is somatic hypermutation restricted to B cells?

A

T cells do not undergo sh as they may lose their ability to recognise MHC

T cells may become self reactive, and would have to be shuttled back to thymus to undergo negative selection again

30
Q

What cells are required for B cell class switching?

A

T follicular helper cells (TFH) required to induce class switching, done by cytokines
– in germinal centres

receptor binding required as well
– CD40L on TFH binds CD40 on B cell

CD40L deficiency leads to inability to class switch and overproduction of IgM

31
Q

How do AID, UNG, and APE1 perform class switching?

A
  1. germline transcript makes RNA:DNA hybrids (R-loops)
    - R loop preferentially acted on by AID
  2. AID converts C bases to U by deamination in switch (S) regions
    - will change multiple Cs in long sequence
  3. Uracil-DNAglycosylase (UNG) will remove U residues leaving abasic site
  4. abasic sites are nicked by APE1 endonucleases
  5. will eventually result in a ds break in S region
    - when this happens in two switch regions, recombination can occur through homology of Switch region sequences (chnage ot IgG, E, or A)
32
Q

How is IgM or IgD produced?

A

differential splicing in mature but naive B cells,

they share same variable region, transcript is spliced to remove exons from m region or d region

33
Q

How are B and T cell receptors used therapeutically?

A

neutralisation - prevent receptor ligation

activation - trigger receptor ligation

ADCC - depletion

antibody-drug conjugates - targeted delivery of treatment

34
Q

What are BiTEs?

A

Bispecific T cell engagers

fusion proteins consisting of two single-chain variable fragments (scFv) derived from different Abs
- one binds to T cells via CD3e
- other binds tumour cell via tumour specific molecule
- this draws two cells closer together and induces T cell signalling via CD3

results in directed tumour lysis and help to drive anti-tumour response

35
Q

What is a CAR T cell?

A

chimeric antigen receptor T cell

Contain a single-chain variable fragment (scFv) derived from an antibody, which binds a tumor antigen (e.g., CD19 in B-cell malignancies)

intracellular CD3zeta signalling domain

recognize antigens independently of MHC
CAR T cells bind to tumor antigens become activated, proliferate, and kill cancer cells through cytotoxic mechanisms

36
Q

What is S1P?

A

sphingosine-1-phosphate

  • a key regulator of T cell trafficking
  • produced by RBCs and lymphatic endothelial cells
  • sensed by a family of 5 receptors (S1PR1 etc)
  • S1PR1 upregulated during thymocyte maturation
  • S1PR1 KO T cells are unable to leave the thymus
37
Q

How is fingolimod used to treat multiple sclerosis?

A

S1P blocker

inhibits T cells from moving out of LN

38
Q

How does S1P signalling work?

A

an S1P gradient drives mature SP thymocytes out of thymus and into the blood

S1PR1 is internalised when it binds S1P
- while in blood, T cells have low surface S1PR1

39
Q

How do naive T cells circulate?

A

recirculate through blood and lymphatics until they meet antigen

40
Q

How do T cells enter lymph nodes?

A

enter cortex from blood through HEVs via adhesion molecules and chemokines

  1. L-selectin (lymphocyte) binds GlyCAM-1 and CD34 allows rolling interaction
  2. LFA-1 activated by CCR7 signalling in response to CCL21 bound to endothelial surface
  3. activated LFA-1 binds tightly to ICAM-1
  4. lymphocyte migrates into LN by diapedesis

due to high S1P binding in blood, there is low levels of expression of receptor
- this allows other signals to easily become dominant process

41
Q

What is the role of the fibroblastic reticular cell network?

A

used by naive T cells to walk around LN and scan DCs
- T cell “random walk”
- if naive T cell does not meet a DC presenting cognate antigen in LN, it goes back into circulation, cycle repeats
– otherwise it establishes a sustained connection w DC

42
Q

How do T cells form the immunological synapse?

A
  1. initial binding through low affinity LFA-1:ICAM-1 interactions
  2. subsequent binding of T cell receptors signals LFA-1
  3. conformational change in LFA-1 increases affinity and prolongs cell-cell contact
43
Q

What 3 signals are required for T cell activation?

A
  1. antigen + co-receptor (CD4/CD8)
  2. co-stimualtion (CD80/CD86 binding to CD28 on T cells)
  3. cytokines which specify differentiation (eg IL-12 for Th1 differentiation)
44
Q

How is downstream signalling of TCR initiated?

A

Lck, a protein kinase, associates w co-receptor (CD4/CD8)

intracellular CD3 chains of TCR have ITAMS (immunoreceptor tyrosine-based activation motifs)

following TCR engagement w MHC:antigen complex
– TCR/CD3 complexes cluster together
– CD3 chains undergo conformational change that makes ITAMS accessible
– CD4-associated Lck is brought into proximity of CD3 to phosphorylate ITAMs
- phosphorylation of CD3zeta creates docking site for ZAP70

– binding of ZAP70 to CD3zeta -> conformational change
– this exposes activating Tyr in ZAP70 which are phosphorylated by Lck
– ZAP70 is now activate and can phosphorylate downstream molecules

45
Q

How does ZAP70 activate downstream molecules?

A

– post activation, ZAP70 phosphorylates LAT -> recruitment of SLP76 via its adaptor, Gads

– the LAT-Gads-SLP76 complex creates a platform for recruitment of phospholipase C gamma (PLCy) and other signalling molecules

46
Q

How does CD28 continue the T cell activation pathway?

A

– CD28 also recruits adaptors and kinases eg PI3K

– PI3K generates phospholipid PIP_3 at PM -> recruits proteins w PH domains

– the kinase Itk is recruited via its PH domain

– Itk activates PLCy

– active PLCy hydrolyses PIP2, which makes IP3 and DAG

47
Q

How does IP3 lead to changes in gene expression?

A

IP3 induces release of Ca2+ into the cytoplasm

this activates calcineurin, which leads to translocation of the TF, NFAT (nuclear factor of activated T cells) into the nucleus

48
Q

How does DAG lead to changes in gene expression?

A

DAG activates protein kinase C theta (PKC theta) which leads to the Ras/MAPK pathway

this results in activation and translocation of TF, AP-1 (activating protein 1)

49
Q

How does PIP_3 lead to changes in gene expression?

A

PIP_3 also activates PDK1, which activates the PKC/Akt pathway

leads to activation and translocation of TF NF-kB (nuclear factor kB)

50
Q

What is the role of CD69 in T cell activation?

A

inhibits S1PR1 by inducing its internalisation
– allows T cell to stay inside LN to allow activation
– once T cells are activated, CD69 is downregulated so it can recognise S1P and migrate out of LN

51
Q

What is the contraction phase?

A

reduction in the number of Ag-specific lymphocytes (only 5-10% remain)

mediated by:
- antigen/growth factor withdrawal
- inhibition of activation/proliferation
- activation-induced cell death

less antigen = less stimulation = less IL-2 = less TLR activation = T cell death

coinhibitory receptors are transiently upregulated after T cell activation to terminate signal eg CTLA-4, PD-1

also Fas/FasL expression on B cells
– also leads to T cell memory development as B cells express FasL

52
Q

How does CTLA-4 inhibit T cells?

A

cytotoxic T lymphocyte antigen 4

– binds CD80/86 w greater affinity than CD28
– outcompetes B7 on APCs
– removes B7 from APCs
– induces inhibitory cytokines eg TGF-beta

inhibitory effects demonstrated by
- agonistic Abs
- phenotype of CTLA-4 KO
– fatal immunoproliferative disease
– reversed by blocking CD28
- Tregs express CTLA-4 constitutively

there is evidence showing Tregs secrete soluble CTLA-4 through alternative splicing

53
Q

How does PD-1 inhibit T cells?

A

programmed death 1

binds PD-L1 and PD-L2
- PD-L1 broadly expressed (vascular, epithelial, hepatocytes etc)
- PD-L2 expressed in APCs

inhibitory effect demonstrated by:
- phenotype of PD-1 KO = increased inflammation
- effect of PD-1 blockade

54
Q

How do coinhibitory receptors signal to achieve inhibition of TCR signalling?

A

recruitment of phosphatases, results in:
- no cell activation
- no induction of pro-survival molecules
- no proliferation

phosphatases important for inhibition
opposite of activation, which require kinases

55
Q

How is AICD carried out?

A

induced by death receptors
- Fas/FasL
- TNFR1
- TRAIL1 and TRAIL2

avoided during clonal expansion by CD28-dependent expression of antiapoptotic factors (cFLIP, BCL-X_i)

subset dependent:
- mouse/human CD4 T cells susceptible to FAS mediated
- human CD8 more susceptible to TNFR1

56
Q

What is AICD and why does it occur?

A

activation induced cell death (also called restimulation-ICD)

effector T cells are susceptible to cell death upon restimulation

After an infection is cleared, AICD helps reduce the population of activated T cells, preventing an overactive immune response

also plays a role in tolerance:
– AICD contributes to the elimination of autoreactive T cells, maintaining tolerance and preventing autoimmunity

57
Q

Why does T cell exhaustion occur?

A

beneficial in chronic infection where response to the infection would be worse than the infection itself

more useful in autoimmune disease, constant exposure to self antigen needs exhaustion to occur

58
Q

What are the differences between active and exhausted T cells?

A

active:
- uses glycolysis and main energy metabolic pathway
- production of many cytokines (polyfunctional activity)
- limited inhibitory receptors
- TCF-1 TF expression associated w activated T cells

exhausted:
- less glycolysis
- reduced proliferative activity
- decreased production of effector cytokines
- reduced cytotoxicity
- increased inhibitory receptors
- change from TCF-1 to TOX expression
- dysfunctional mitochondria (increased ROS)

59
Q

How can T cell exhaustion be inhibited for cancer immunotherapy?

A

ipilimumab: anti-CTLA-4
nivolumab: anti-PD-1
pembrolizumab: anti-PD-1

60
Q

What are the 3 signals that lead to T cell exhaustion?

A
  1. persistent antigen
  2. negative costimulation
  3. chronic inflammation
    – mitochondrial stress
    – metabolic byproducts eg lactic acid
    – cytokine dysfunction
    – Histone modifications and DNA methylation at genes associated with effector functions limit the ability of T cells to regain their activity
61
Q

How is the exhaustion pathway thought to work?

A

exhaustion has been studied in CD8 T cells more sure role in cancer immunity, but idea is thought to be same in CD4 too

thought to be a pathway of gaining certain inhibitory receptors in sequence, gradually becoming more and more exhausted until they are susceptible to apoptosis

62
Q

What are some unanswered questions regarding T cell exhaustion?

A

is it reversible?

is exhaustion more fixed the more exhausted it is?

differences in process between chronic infection and tumours?

can epigenetic changes be reversed?

63
Q

What are progenitor exhausted T cells?

A

Progenitor exhausted T cells (Tpex)

identifiable by high levels of Tcf1, Tox and Ly108 expression, have been described as a source for the effector-like T cells in settings of chronic viral infection and cancer

their quantity is preserved through Tpex undergoing self-renewal, which provides a cellular mechanism for their long-term maintenance, “stem-like”

64
Q

What is anergy in the context of peripheral tolerance?

A

a state of unresponsiveness from stimulation from an unactivated APC
– occurs when APC presents self antigen (APC is not activated as it is self antigen), and T cell goes into anergy (as it recognises self)

no cytokines produced eg IFNy, IL-12
no co-stimulation

65
Q

How is anergy induced?

A

DAG activates protein kinase C theta (PKC theta) which leads to the Ras/MAPK pathway
-> activates AP-1

in the absence of AP-1, NFAT induces expression of anergy genes such as DGK (a kinase) which breaks down DAG
- maintains anergic state

reminder: DAG is recruited and activated as a result of CD28 binding
– no costimulation = no DAG = anergy

however, if there is not costimulation, then there wouldnt be any DAG recruitment, so why would DAG need to be degraded?

66
Q

How is apoptosis induce in peripheral tolerance?

A

deletion of autoreactive or potentially dangerous T cells

induced by mitochondrial pathway:
- deletion of T cells stimulated in tolerogenic conditions
– tolerogenic conditions refers to immunosuppressive conditions or absence of danger signals

  • regular TCR stimulation leads to expression of proapoptotic molecules (eg BIM), but in normal signalling (Cd28 + cytokines), survival factors counteract these molecules (eg BCL2)
  • absence of costimulation = no survival factors = apoptosis
67
Q

What mechanism determines whether anergy or apoptosis occurs?

A

Anergy:
– Typically induced by weak or transient TCR signalling in the absence of co-stimulation
– Low-affinity interactions with self-antigens or insufficient engagement of the TCR fail to provide the robust signal needed for full activation or apoptosis, leading to a hyporesponsive state

Apoptosis:
– Strong, prolonged TCR signalling without adequate co-stimulation or in the presence of inhibitory signals can trigger apoptotic pathways
– Chronic or repeated exposure to the same antigen (as in chronic inflammation) often results in apoptosis.

68
Q

What is the difference between apoptosis and AICD in the context of peripheral tolerance?

A

AICD is the deletion of a T cell that has already been activated

69
Q

How does ALPS demonstrate the role of apoptosis in peripheral tolerance?

A

autoimmune lymphoproliferative syndrome

occurs in patients w mutations in apoptosis pathway eg Fas, FasL, Caspase 10

70
Q

What are iTregs?

A

CD4+ T cells that have been induced to become Tregs in the periphery

arise during activation in the presence of TGF-beta, which induces Foxp3 expression (Treg master regulator)

71
Q

How do Tregs (i or n) inhibit T cell function?

A

iTregs constitutively express CD25 and CTLA-4
– v high expression of CD25, binds all environmental IL-2, inhibiting T cells

induce DCs to secrete IDO, which is a suppressive signal for effector T cells

Tregs express CD39 and CD73 (enzymes) on PM
– CD39 converts ATP and ADP -> AMP
– CD73 further degrades AMP -> adenosine
T cells express adenosine receptor A2AR, binding results in:
– reduced pro-inflammatory cytokine production
– glycolysis to oxidative phosphorylation, reducing the energy available for rapid T cell proliferation and function