Autoimmunity (Rheumatoid Arthritis) Flashcards

1
Q

What is the issue with the adaptive immune system?

A

• Adaptive immune system has high diversity due to somatic recombination of VDJ segments in lymphoid organs, and somatic hypermutation (of BCR) in peripheral organs.
− Adpative provides capacity to recognise any chemical structure → BUT! Potential for autoimmunity!

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

Why is an effective immune system a compromise?

A
  • Need removal of dangerous self-reactivity but without impairing the capacity for broad recognition and effective defense
  • If removal were too rigorous – holes in the repertoire
  • Some self-reactivity is normal – limited by mechanisms of tolerance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the 3 mechanisms of immunological tolerance?

A
  • Inactivation of potential self-reactive clones → death, receptor editing, anergy
  • Immune regulation → suppression or functional deviation
  • Antigenic epitopes not available for recognition → ignorance, sequestration, immune privilege
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Describe central tolerance in B cells

A

• No self reaction:
− Immature B cells that don’t have strong reactivity to self antigen allowed to mature. Leave the bone marrow and carried to the spleen.
• Clonal deletion/receptor editing:
− Strong, multi-valent cross-linking by self antigen
− Interval before cell death where the B cell may be rescued by further gene rearrangement reaplcing the autoreactive receptor with one that isn’t.
− Upon strong cross-linking, self-reactive light chain is deleted and RAG expression continues, with light chain-rearrangement continuing until a non-autoreactive receptor produced
− Cells that remain autoreactive when all V and J segments are exhausted undergo clonal deletion by apoptosis.
• Anergy:
− Weakly cross-linking self antigen with low valence
− B cells enter a state of permanent unresponsiveness
− Anergic B cells cannot be activated by their antigen, even with T cell help
− Migration of anergic B cells within lymphoid organs is impaired (they are excluded from follicles), and they cannot compete with immunocompetent B cells
− Without T cell survival signals, they eventually die
• Clonal ignorance:
− Have affinity for self antigen, but do not sense and respond to it
− Antigen may not be accessible, may be in low concentration, or may bind so weakly that it does not generate an activating signal
− Some ignorant cells are activated under certain conditions such as inflammation or when antigen becomes available and reaches an unusually high concentration – so they should not be considered inert.

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

Describe the 3 checkpoint stages of central tolerance in T cells

A

Checkpoint 1: Quality check → Cortex
• Has the TCR-beta gene rearrangement resulted in a functional pre-TCR expression?
• Occurs at the DN3 stage
• If it hasn’t, the cell fails to receive a survival signal and is deleted by apoptosis

Checkpoint 2: Positive selection → Moving towards the medulla
• Can the rearranged abTCR recognise self MHC?
• DP thymocytes encounter MHC-I or MHC-II on the surface of cTECs
• Thymocytes who can bind this antigen receive a survival signal
• Cells that cannot bind can be recovered by receptor editing, or die by apoptosis
• Only 10-30% make it to the next stage

Checkpoint 3: Negative selection → Medulla
• Does the abTCR recognise self-MHC and peptide too strongly?
• Antigen presented by mTECs
• T cell clones deleted if they bind too strongly

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

What presents antigen in the thymus for central T cell tolerance?

A

cTECs:
• Constiutive MHCI and II expression
• MHC 1 peptides → processed by thymoproteasome
• MHC II peptides → processed by
− Cathepsin L in the endosome
− Thymus specific serine protease (TSSP) in the endosome or lysosome
− Constitutive macroautophage

mTEC:
• Constitutive MHC I and II expression
• MHC I peptides → processed by the housekeeping proteasome and immunoproteasome
• MHC II peptides → processed by capthepsin S and macroautophagy

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

What are the sources of antigen for central T cell tolerances?

A

mTECS:
• They are inefficient at uptake and presentation of exogenous antigen
• They therefore express AIRE → allows for promiscuous expression of tissue specific genes not normally expressed in the thymus
• Genetic deficiency of AIRE results in autoimmune polyendocrinopathy
• mTECS also perform macroautophagy, so their cellular contents can be degraded and expressed on MHCs

DCs:
• Uptake tissue restricted antigens shed form mTECs
• Uptake of blood-bourne antigen
• Cross-presentation of ingested antigen

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

Summarise the affinity hypothesis of T cell selection

A
  • T cells that fail to make a functional TCR → die at checkpoint 1
  • TCR that cannot recognise self-MHC → die at checkpoint 2 – positive selection
  • TCR that recognizes self-MHC and self-antigen too strongly → die at checkpoint 3 – negative selection
  • TCR that recognizes self-MHC and doesn’t bind self antigen too strongly → good
  • T cells that recognise self-MHC and bind self-antigen moderately (not too strong that they are deleted) → become Foxp3+ TRegs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Describe peripheral tolerance in B cells

A

• Mainly due to lack of T cell help

  1. DC sees an antigen in the periphery, presents it and migrates to the lymph node
  2. Activates a T cell in the lymph node
  3. Activated T cells move to the B cell region of the lymph node
  4. Some of the antigen will have drained to the lymph node, and been taken up and presented by B cells
  5. If the T cell sees that the B cell is presenting the same antigen as the DC → gives it costimulation to become a plasma cell
  6. If however, a self antigen had arrived at the lymph node, the B cell would have presented it but there would be no T cell help, because a DC would not have recognized the antigen as dangerous, so wouldn’t have activated the T cells.

Another thing to consider is activation in germinal centres:

  1. Competition for limited antigen and Tfh cells drives selection for highest binders
  2. Failure to bind and present antigen = failure to receive survival signals
  3. Binding of soluble self antigen = apoptosis
  4. T follicular regulators = antigen specific suppression of B cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Describe peripheral tolerance in T cells

A

• Antigen must be presented on a DC in an activation context to initiate naïve T cell activation
• Presentation in the absence of danger (without signal 2 or 3) is tolerogenic → leads to energy or regulation
• Clonal anergy:
− Presence of signal 1 without 2 or 3 renders a T cell anergic
− It is unresponsive to antigen – doesn’t turn into an effector cell
• Regulation → peripheral induced TReg
− Presence of signal 1 with immunosuppressive signal 3 skews the T cell to become a Treg, rather than an effector
− Number of different TRegs depending on the cytokine that is signal 3
− TGFB → Foxp3+ iTreg
− IL-10 → Tr1 (Foxp3-)

How regulatory T cells regulate:
• Production of anti-inflammatory cytokines → TGFB, IL-10
• Expression of inhibitor receptors → CTLA-4
• Modulation of DCs
• Outcompete effector T cells for resources → CD25 mops up IL-2 (acting as a cytokine sink)
• Direct killing of T cells using granzyme and perforin.

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

What is autoimmunity?

A
  • Loss of tolerance to self antigen, resulting in specific adaptive immune responses to self-antigen.
  • Loss of central tolerance likely occurs all the time, but is normally kept in check by peripheral tolerance
  • It may or may not cause disease → you can have an autoimmune response resulting in autoantibodies that circulate for years without causing disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is autoimmune disease?

A
  • Tissue response and damage resulting from autoimmunity
  • Self antigen cant be cleared, resulting in chronic pathology
  • Results from the failure of peripheral tolerance and dysregulated activity of normal immune effector functions → there is nothing new or overactive, it is just dysregulated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What may contribute to B cell-mediated loss of tolerance and autoimmune disease?

A

− Non-specific, T cell independent activation of B cells by microbial mitogens, eg) LPS, DNA and other TLR ligands

− molecular mimicry → T cell dependent. Sequence similarities between foreign and self-peptides are sufficient to result in the activation of autoreactive T or B cells by pathogen derived peptides.
− eg) Rheumatic fever
➢ Immune response to Streptococcal cell surface M-antigen generates antibodies that cross-react with self-glycoproteins in the heart, skin and connective tissue
➢ In susceptible individuals (doesn’t happen in everyone who gets infected), this gives rise to autoimmune disease where tissue damage is mediated by complement, neutrophils and macrophages

− genetic variation associated with altered signaling pathways
− eg) altered activation thresholds and ineffective negative selection
− or to enhanced germinal centre activity (e.g. somatic hypermutation leading to auto-reactivity) and preferential selection by autoantigens during the generation of memory B cells and plasma cells.

− Cell-type specific epigenetic changes resulting in altered gene expression in cells of the immune system.

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

What may contribute to T cell-mediated loss of tolerance and autoimmune disease?

A

− impaired generation of TRegs
− post-translational modification of self-proteins or altered availability providing neo-antigens
− Altered availability: may have tissue damage resulting in antigen release from immune privileged sites. T cells wont have seen these during central tolerance
− Altered self:
➢ Post-translational modification, eg) citrullination of Arg residues implicated in RA
➢ Chemical modification, eg) oxidation of type II collagen in RA joints
− Molecular mimicry: foreign antigen and self protein have similar epitopes, and be taken up and presented by APC. The T cell generated against the foreign antigen may be able to react to self antigen.
− genetic variation associated with altered signaling pathways
− Cell-type specific epigenetic changes resulting in altered gene expression in cells of the immune system.

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

What causes the transition from autoimmunity to chronic autoimmune disease?

A

Early:
Activation phase:
• May have some initial tissue damage or cell death
• Autoreactive B and T cells respond to autoantigen that is released, and produce autoantibodies
• This may occur at relatively low levels

Later:
Chronic phase:
• Further tissue damage occurs
• This breaks down sequestration
• Autoantibodies bind self-antigens and non-specific effectors (macrophages, neutrophils) are attracted
• Release of inflammatory mediators
• Self-destructive process perpetuated
• Epitope spreading
− refers to the development of an immune response to epitopes distinct from, and noncross-reactive with, the initial activating epitope
− Thee ability of the immune system to attack multiple targets on a pathogen has obvious advantages, but it can result in escalating autoimmunity.
− Epitope spreading in diabetes:
➢ Initial responses are towards central epitopes of glutamic acid decarboxylase (GAD)
➢ As the damage progresses, the response spreads to other GAD epitopes → intramolecular epitope spreading
➢ The response may also spread to other antigens, eg) insulin, HSP, carboxypeptidase H → intermolecular epitope spreading
− Epitope spreading in SLE:
➢ Nucleosomes may be recognized by a B cell, taken up, processed and antigen preented to a T cell
➢ This could result in antibodies against the histone or the DNA component
➢ Tend to see histone first, then DNA later

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

Describe the pathology of autoimmune diseases

A
  • Primary pathology → a direct consequence of the autoimmune response
  • Secondary pathology → arises as a consequence of the primary pathology

eg)
T1DM:
• Primary → autoimmune destruction of pancreatic beta cells
• Secondary → metabolic effects due to lack of insulin

Hashimotos thyroiditis:
• Primary → destruction of the thyroid tissue
• Secondary → hypothyroidism – causes fatigue, intolerance of cold, dry skin, mental impairment

• There is organ specific and organ non-specific disease
− Organ specific → eg, Hashimotos thyroiditis
− Organ non-specific → eg, SLE, RA

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

What are the immunological features that cause autoimmune disease? (T Cells/Autoantibody)

A
•	Autoantibodies
−	In the serum
−	Deposited in tissues
•	Cellular infiltrate
−	T cells, B cells, Monocytes, NK cells, DCs, Neutrophils 

• Most autoimmune diseases involves the generation of autoreactive CD4+ T cells
− These drive other immune effector functions (B cells and CTLs) and recruit and amplify innate effector cells
• However, in some conditions, autoantibodies are the primary mediators due to:
− Complement mediated lysis
− eg) Autoimmune haemolytic anaemia
− Anti-RBC autoantibodies bind to RBCs. Complement is fixed and leads to generation of the MAC. Results in lysis of the RBC
− Opsonisation
− Eg) Autoimmune thrombocytopenia
− Anti-platelet autoantibodies binds to the platelet, acting as an opsonin, leading to phagocytosis of the platelets
− Antibody mediated stimulation of function
− Eg) Graves disease (hyperthyroidism)
− In a normal situation, the pituitary secretes TSH, which acts on the thyroid to secrete thyroid hormone. These then act back on the pituitary as negative eedback, suppressing further TH synthesis.
− In Graves disease, autoantibodies against the TSH receptor stimulate thyroid hormone production
− Antibody mediated inhibition of function
− Eg) Myasthenia gravis → muscle weakness caused by inhibition of signals to the muscle
− Antagonist → autoantibodies blocks binding to the nicotinic acetylcholine receptor
− Removal of receptor → autoantibody triggers receptor internalization
− Antibody mediated deposition of immune complexes
− Eg) SLE → symptoms include butterfly rash, arthritis, vasculitis, glomerulonephritis
− Immune complexes deposit in small blood vessel walls (kidney, joints, skin) and initiate inflammatory reactions

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

In autoimmune disease, autoantibodies against nuclear antigen are prevalent - describe some features of this.

A
Antibodies against nuclear antigen:
•	Particular prevalent in SLE, where almost all patients have autoantibodies against nuclear antigens, but seen in other diseases and also healthy controls (highlighting the fact  you can have autoantibodies without disease)
−	SLE → 99% of patients
−	Scleroderma → 80%
−	RA → 50%
−	Relatives of SLE patients → 20%
−	Healthy controls → 5%
•	Antigens include:
−	dsDNA – renal disease – 40-60% prevalence
−	Ribonuclear protein – connective tissue disease – 30-40%
−	Phospholipids – thrombocytopenia – 30%
−	Histones – SLE

• These antigens may be exposed due to impaired clearance following apoptosis and NETosis (apoptosis that occurs in neutrophils. Can contribute to phagocyte clearance but releases the contents of the neutrophil into the periphery)
− Usually, apoptotic cells are silently cleared by complement, but in SLE this is defective

As stated before, high affinity autoantibody generation may also arise because of intrinsic B cell defects due to genetic variants.

Overexpression of IFNa and BAFF anti-nuclear antibody production in SLE:
• Genetic variant of the FcyRII – can influence the ability to recognise immune complexes
• When plasmacytoid DCs take up the nuclear DNA/antibody immune complex, it triggers the production of type I IFN
• This can activate monocytes and DCs to produce BAFF – present at very high amounts in SLE patients due to altered gene expression control
• Can result in loss of negative feedback loop that would normal result in death of autoreactive B cells and can enhance their survival

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

What is the concordance rate in autoimmune disease?

A

In autoimmune disease:
• Concordance in identical twins > non-identical, but concordance in identical twins is 50% → genetic and environmental factors

eg) In RA:
• Unrelated individuals → 0.5-1%
• First degree relatives → 3-5%
• Identical twins → 15%

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

Other than concordance, what else indicates an environmental influence in autoimmune disease, and what are potential triggers?

A
  • Rapid increase in incidence over the last 40-50 years too quick to be explained by genetics
  • Geographical movements – populations moving from areas of low to high incidence reflect the incidence of the new location.

Possible environmental triggers:
• Infections → microbial pathogens may resemble self-antigen
• Drugs → may bind to self-epitope, yielding a neoantigen
• Toxins (UV, pesticides, smoking) → cause damage to tissues, expose sites for autoantibody mediated damage, give rise to protein modifications and neoantigens
• Diet → Through alterations of epigenome and effects on gut microbiome
• Obesity → adipokines are proinflammatory
• Hygeine → lack of challenge to immune system by parasites
• Vaccines → due to adjuvants

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

Name some single gene deficiencies giving rise to autoimmune syndromes.

A

AIRE
Autoimmune polyendocrine syndrome. T-cells escape negative selection due to decreased expression of self-antigen in thymus

Foxp3
IPEX . Decreased generation of TRegs

FAS
Autoimmune lymphoproliferative syndrome. Failure of apoptosis of self-reactive T and B cells

C1q
SLE. Failure to activate classical pathway of complement, leading to poor immune complex clearance

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

Other than single gene deficiences, what other general genetic associations have been found for autoimmune disease

A

Sex-related genes are also a risk factor:
• 75% autoimmune diseases occur in females
− SLE – 9:1 female to male
− Graves – 7:1
− Arthritis – 2:1
− Ankylosing spondylitis – 2:1 male to female
• Symptoms usually arise during childbearing years – symptoms can be altered during pregnancy

Most autoimmune diseases are polygenic:
• Susceptibility influenced by genetic variation
− Eg) naturally occurring SNPs , not mutations
− May directly affect protein function, or influence gene expression and hence cell phenotype
− Some conditions associated with allelic variants in large numbers of genes
− >30 genes contribute to SLE
− >40 contribute to RA
• Individual variants account for only a small proportion of genetic risk
− Each make a subtle contribution to dysregulation of immune function
− Requirement of allelic combinations (epistasis) to confer substantial risk
− epigenetics , which can be altered by the environment, can contribute
− non-coding transcripts, eg, miRNA can also influence gene expression
• Some gene variants affect susceptibility to multiple diseases, suggesting common pathways underlying the aberrant immune response
− eg) disease associated variants of PTPN22 and CTLA4 modulate the signaling pathways of T cells

Influence of MHC polymorphism:
• Strongest genetic associations are with MHC alleles (usually class II) → often 50% of genetic risk
− HLA-DR3 predisposes for Graves, HLA-DR7 is protective

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

How have GWAS been used to identify genetic factors in autoimmune disease?

A

• Scan the whole genome for SNPs associated with autoimmune disease
− 100s loci identified, many containing non-coding transcripts and regulatory elements
− 2/3 of these influence multiple conditions, suggesting effects on common pathways such as cytokine signaling and antigen presentation
• New algorithm PICS used to fine-map SNPs associated with 21 autoimmune diseases, in conjunction with epigenetic mapping to determine the active elements:
− 90% of causal SNPs are non-coding
− 60% map to immune cell enhancers, many of these CD4 enhancers that undergo acetylatio

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

What are the effects of genetic variants on immune responses in SLE

A
Effects of genetic variants on the immune response in SLE:
•	Processing and clearance of immune complexes → environmental triggers induce apoptosis and release of nuclear antigen. Defective clearance is a feature of SLE
−	C1q, C2, C4
−	FCGR
−	TREX1
•	TLF-IFN signaling → UV, drugs, viruses can yield stimulatory DNA or RNA that activates TLRs, resulting in type I IFN secretion
−	TLRs
−	IRANK
−	STATs
−	IFN
•	Signal transduction to the adaptive immune response → presentation of nuclear antigens by DCS leads to autoantibody generation and immune complexes
−	HLA
−	BCR
−	TCR
−	STATs
−	PTPN22
−	Cytokines
25
Q

What is RA and what are the general features?

A
  • Chronic, systemic, inflammatory autoimmune disorder mainly affecting articular joints
  • Common in both human and nimals
  • May have something to do with joints having to provide lots of support to the body making them particularly susceptible to inflammation
  • Autoimmunity is initiated at mucosal sites when self proteins are modified in response to environmental stimuli
  • Disease-specific autoantibodies can be detected in the sera several years before clinical manifestation = pre-arthritis phase. In some, they may have these for years and never go on to develop RA
  • Inflammation of the synovium (synovitis) occurs during early clinical disease
  • In advanced clinical disease, destruction of the synovial joints leads to severe disability
26
Q

What are the clinical symptoms of RA?

A

Early → due to active inflammation
• Morning stiffness of affected joints
− Prominent feature, typically lasts more than an hour
− Gentle movments may relieve symptoms – helps distinguish rheumatoid from non-inflammatory osteoarthritis, as in non-inflammatory conditions morning stiffness is less prominent and movement induces pain
• Generalised fatigue, anaemia

Late:
• Pain, swelling, loss of mobility of affected joints due to cartilage damage, tendon tethering and bone erosion
• Typically effects:
− Wrists and fingers → ulnar deviation, boutonneire deformity, swan neck and Z-thumb
− Shoulders
− Elbows
− Hips
− Knees
− Ankles
• Increased susceptibility to cardiovascular disease and osteoporosis longer term
• Other non-joint features:
− The rheumatoid nodule, sometimes in the skin, is the most common non-joint feature
− Occur in 30%
− Inflammatory reaction known as a necrotizing granuloma
− Central area of fibrinoid necrosis corresponding to the fibrin rich material found an in affected synovial space
− Surrounding this is macrophages and fibroblasts
− Maybe a few mm to a few cm in diameter, usually over bony prominences such as elbow, heel or knuckles
− The eye is directly affected in the form of episcleritis
− The chronic inflammation caused by RA leads to raised hepcidin levels, leading to anaemia of chronic disease where iron is poorly absorbed and sequestered into macrophages
− Peripheral neuropathy and mononeuritis multiplex may occur – most common problem is carpal tunnel syndrome caused by compression of the median nerve by swelling around the wrist.

27
Q

What is a synovial joint, and what happens to it in RA?

A
  • Joins two opposing bones with a fibrous joint capsule continuous with the periosterum of the joined bones
  • Covered in cartilage, providing load bearing properties
  • The synovial cavity is filled with synovial fluid – has a small number of cells to provide nutrition and protect the joint
  • In RA, the synovium is inflamed and expanded. Cartilage has been damaged, resulting in reduced joint space. Can also see damage to the underlying bone as the disease progresses
  • RA usually affects joints on both sides equally
28
Q

Give an overview of the inflammatory processes in RA.

A
  • Chronic, unresolved inflammation in the synovial joints is assocated with pannus formation (a pannus is a layer of vascular fibrous tissue extending over the surface of an organ or specialized anatomical structure), neo-vascularisation and lymphocyte accumulation.
  • Activation of chondrocytes and differentiation/activation of osteoclasts leads to progressive cartilage destruction and bone erosion, hence disability.
  • Inflammatory mediators inc. cytokines and immune complexes circulate to promote pathogenesis in other organs.
29
Q

How is RA diagnosed?

A

• Usually occurs when the patient goes to the doctors with unexplained synovitis in at least 1 join
• The criteria is not diagnostic, but a classification criteria to identify disease with a high likelihood of developing a chronic form. However, a score of 6 or more unequivocally classifies a person with RA.
• Criteria based on:
− Joint involvement, ie, swelling, tenderness, pain (1 joint is 0 points, 4-10 joints 3)
− Serology
− Important for determining if there is autoimmune disease
− Looks for rheumatoid factor (antbodies to the Fc region of self IgG)
− Looks for anti-citrullinated peptide autoantibodies (ACPA)→ 70% of cases are ACPA+
− Negative is 0 points, high positive is 3
− Acute phase reactants
− Abnormal CRP and/or ESR
− Duration of symptoms
− More or less than 6 weeks

30
Q

What is the prevalance of RA?

A
  • 2-3 times more women than men
  • Age of onset is typically 35-50 years (osteoarthritis on the other hand tends to onset later)
  • Overal UK prevalence is 1%
  • Data indicate prevalence has fallen in women by 20-30% over the last 50 years, but not men
  • Prevalence is lower in developing countries, and appears lower in Southern Europe than Nothern (maybe due to different in Vitamin D levels?)
31
Q

What are the potential mechanisms for the breakdown of tolerance in RA?

A

Antibody crossreactivity:
• B cells that express ACPAs and recognise citrullinated epitopes on pathogens could receive help from CD4+ T cells that respond to citrullinated-pathogenic peptide complexes, resulting in the secretion of ACPAs

T-cell crossreactivity:
• Molecular mimicry could result in loss of T cell tolerance, where T cells activated in response to pathogenic citrullinated peptides could cross-react with self-citrullinated epitopes due to high levels of similarity
• Self reactive T cells could provide help to B cells that recognise citrullinated proteins

Generation of post-translational modifications:
• Modificaiton, eg, citrullination, due to genetic and/or environmental factors (often occurs in the context of inflammation) could result in neoepitopes
• This could result in activation of T cells that were not negatively selected against in the thymus, because those epitopes weren’t available during central tolerance

→ So, ACPAs may be against normally occurring citrullinated peptides as a result of molecular mimicry and a loss of tolerance, or post-translational modifications generating neoepitopes.

How?
• Citrullination is mediated by PAD enzymes produced by apoptotic cells or in inflammation
• Positively charged arginine is converted to neutral citrulline – creating a neoepitope
• Citrullinated proteins are found at inflammatory sites, eg) lungs of smokers and in chronic periodontal disease – both these are associated with RA
• They are also found in the joints of RA patients and in other forms of arthritis, but ACPAs are 95% specific to RA.

ACPAs
• Found in the sera of 70% of RA patients
• Can be detectable up to 10 years before the onset of symptoms
• Potentially pathogenic, but their presence does not cause RA, something else needs to happen

32
Q

ACPAs dont CAUSE RA - so how does the breakdown of tolerance lead to pathology?

A
  • Levels of ACPA rise during the pre-clinical stage and change their characteristics → increased avidity, epitope spreading, altered isotype usage, altered glycosylation
  • This leads to increased affinity for Fc receptors
  • This influences effector functions
33
Q

Are there RA specific autoantigens?

A

• In diagnostic tests, ACPAs are detected using synthetic cyclic citrullinated peptides, but doesn’t tell us about the in vivo situation
• Recent studies have shown that cFibrinogen, cVimentin, type II collagen and cTenascin C:
− Are present in the joints of RA patients
− contain defined citrullinated epitopes that are recognized by specific ACPAs → some of these contain dominant epitopes as well as additional epitopes, consistent with epitope spreading
− Immune complexes of ACPA with cFIb shown to stimulate production of TNF by macrophages
− Antibodies specific for cVim promote differentiation of osteoclasts, leading to erosion

34
Q

Describe the HLA-DRB1 association in RA - general features. What is the shared epitope? What is the molecular basis?

A

HLA-DRB1 Association:
• The best characterized association
• 78% Caucasian RA patients had the HLA-DRw4 allotype compared to 28% of controls
• An animal acid sequence (QKRAA) in the alpha helix (residues 70-74) of the DR-beta chain was identified as being a feature of disease-assocaited alleles → THE SHARED EPITOPE
• Individuals with disease associated HLA-DRB1 alleles have a 4-6 fold increased risk of RA

The ‘shared epitope’:
• Residues 71 and 74 contribute to the peptide binding groove
• Lysine at 71 and alanine at 74 polymorphisms are particularly assocaited
• The hypothesis is that the shared epitope alleles may predispose RA via:
− Positive selection of auto-reactive T cells in the thymus?
− Preferential presentation of arthritogenic peptides (neoepitopes) to effector T cells?
− Failure to promote generation of TRegs?
− Facilitating molecular mimicry by microbial proteins?
− Pro-inflammatory signaling?
− As well as residues increasing risk, others are protective
• Serine at position 11 gives almost 25% reduced risk, whereas valine increases risk

Molecular basis for the association of HLA-DRB1 with RA:
• The shared epitope is strongly associated with the presence of ACPAs specific for cTNC, cVIM and cFIB
• The positively charged pocket of RA-associated alleles preferentially accommodates citrulline rather than arginine
• Citrullination altered the protease susceptibility of vimentin, resulting in the production of self-epitopes that could be presented to T cells via RA-associated DRB1 alleles
• RA patients with the HLA-DRB1*0401 allele show:
− Autpreactive T cells specific for citrullinated peptides of correlating with disease activity
− Deficiency in TRegs

35
Q

What are the other HLA associations with RA?

A

Other HLA associations:
• HLA-B has also been implicated → associations with both MHC-I and II suggests both CD8+ and CD4+ T cells contribute to pathology
− Asp-9 in the peptide binding groove of HLA-B*08 allotype is in linkage disequilibrium with other markers in the MHC, eg) those in the TNF region
− So, may not be that the class I molecules is involved in RA, may be a nearby gene – linkage disequilibrium makes it difficult to know what the precise factor is that is the risk.

36
Q

How might the shared epitope behave as a signalling molecule?

A

The shared epitope may also act as a signaling molecule:
• The shared epitope binds to calreticulin on immune cell surfaces
• In DCs, this can mediate signaling pathways that polarize to Th17 differentiation
• IL-17 contributes to RA disease pathogenesis → express high levels of RANKL which promotes osteoclast activity
• Evidence for this came from:
− Mice transgenic for the human shared epitope develop more severe collagen-induced arthritis than wild type, with increased osteoclast numbers
− A cell-free shared epitope peptide mediated increased osteoclast activity in the absence of any antigen presenting activity
− A specific binding site for the shared epitope has been mapped to calreticulin

37
Q

Other than HLA, what are other RA susceptibility genes?

A

• PTPN22:
− Protein tyrosine phosphatase regulating lymphocyte activation – downregulates T cell activation
− RR of 1.8 in the European population
− The PTPN22 Arg-620→Trp-620 confers susceptibility
− Conflicting evidence as to whether its altered function is gain or loss of function
− The effects may differ in different T cell populations
− May allow high affinity, self-reactive TCR expressing T cells to escape negative selection
− Could cause deficient regulation of autoreactive T cells in the periphery
• PADI4:
− PAD enzyme that converts arginine to citrulline
− Citrullinated peptides have increased affinity for the shared epitope
− This polymorphism explains why some people are more prone to citrullination than others
• CCR6:
− Expressed by Th17 cells – correlated with presence of IL-17 in patients sera
• STAT4
− Involved in cellular responses to IL-2, IFNs – can influence the effector phenotype of T cells, eg, promote Th1
• TNFAIP3
− Encodes a negative regulator of NFkB, regulates TNFa production
• CTLA-4
− Downregulates CD28-mediated co-stimulation

38
Q

What are the environmental risk factors for RA?

A

• Smoking:
− In a study of 13 monozygotic twin pairs, it was the smoker who developed RA in 12/13 cases
− Strong association between smoking and ACPA+ RA in patients with the HLA-DRB1
− Other airway exposures, eg) infections and silica dust can trigger RA
• Microbiome
− Factors such as changes in diet or use of antibiotics can alter the balance of the microbiome leading to activation of the innate immune response
− In pre-disposed individual, this could contribute to disease initiation
• TLR activation
− Stimulation by DAMPs or PAMPs leads to activation of innate immune response
− Can lead to expression of PAD enzymes and hence citrullination and neo-antigens
− TLR2 and TLR4 are expressed in the synovium – may prime joints to be more susceptible to RA, and will activate APCs to secrete inflammatory cytokines
• Viral infection
− Potential association between RA and EBV and human herpes virus (HHV)-6.

39
Q

What is the role of mucosal sites in the development of RA?

A
  • In established RA, plasma cells in the synovium generate autoantibodies, but the synovium is unaffected in preclinical RA
  • Patients can have RA-specific antibodies for years without synovial inflammation
  • The initial inflammation/autoimmune response occurs outside the joint.
  • Inducible MALT can develop as a result of chronic inflammation, this can include plasma cells producing ACPAs

Evidence for the role of mucosal sites:
• Environmental factors that affect the mucosa are predisposing to RA
• The predominant antibody isotype at mucosal sites is IgA, and the ACPAs found in early RA are IgA

Periodontal disease:
• PD due to infection strongly linked to RA
• RA and PD known to share pathogenic mechanisms of bone and tissue destruction
• RA related autoantibodies elevated in patients with PD
• Inflamed gingival tissue contains increased levels of PAD and citrullinated proteins
− P.gingivalis is the only known prokaryote with a gene encoding for PAD
− Anti-P.gingivalis antibodies correlated with the presence of ACPAs
• Treating PD appears to improve RA severity and decrease pro-inflammatory biomarkers
• RA patients with active PD are less responsive to treatment

Lung inflammation:
• Increased risk of RA with inhaled exposures eg) smoke, textile tissue
• Inflammatory airway disease associated with RA-related antibodies
• Lung disease can precede clinical RA
• ACPAs detected in the sputum of individuasl without RA and without ACPA in serum – indicating they can be generated in the lungs

The gut microbiome:
• As the gut represents the largest concentration of immune cells, makes sence that microbial make up of an affected individual could influence autoimmune disease
• The toxemic factor hypothesis was formulated at the turn of the 20th century – proposed an abundance of gram-negative bacteria lead to increased toxic metabolites that entered the circulation and ultimately promoted joint inflammation.
• In dysbiosis, rather than promoting TReg formation, microbes can cause expansion of pro-inflammatory T cell populations and promote B cell differentiation.
− Plasma cells and ACPAs migrate to the synovium where the inflammatory cascade is amplified through the activation of macropahges, fibroblasts, osteoclasts and elevated production of cytokines and proteinases.
− Leads to arthritis and pannus formation
➢ RA patients showed overall fewer bacterial counts
➢ RA patients shown an increased diversity of the Lactobacillus communities in early disease

ACPAs and the synovium
In ACPA+ patients:
• 0.005% B cells in the blood produce ACPA
• 10-20% B cells in the synovium produce ACPA
• IgA most abundant isotype in synovial fluid – suggestins an ongoing B cell response to citrullinated peptides
• Ectopic lymphoid structures have been idenfied in the synovium of some RA patients – these can support germinal centre function (B cell differentiation, class switch, secretion of ACPAs)

40
Q

What are the epigenetic factors associated with RA?

A

• DNA methylation at CpG dinucleotides in regulatory regions of genes can repress transcription by inhibiting transcription factor binding
− Altered methylation of the CLTA-4 promoter has been linked to reduced CTLA-4 expression
• Histone modification – by acetylation and methylation – can result in chromatin remodeling and altered gene expression
o In RA synovial cells, specific methylation/acetylation patterns have been implicated in gene expression profiles that promote proliferation and inhibit apoptosis – hence pannus formation.
o Meta analysis of GWAS data (2014) showed an enrichment of H3K4me3; associated with active promoters/enhancers) in RA risk alleles, particularly in Treg cells.
• X chromosome inactivation in females is associated with gene silencing via high levels of DNA methylation and low levels of histone acetylation. The RA susceptibility gene IRAK1, which is on the X chromosome, escapes inactivation in humans.
o A possible epigenetic mechanism contributing to the sex bias in RA.
• miRNAs negatively regulate protein translation via antisense RNA-RNA interactions.
o Dysregulated miRNA expression (e.g. miR-155, miR-146a) promotes protease and cytokine production by RA synovial cells leading to inflammation and matrix breakdown.

41
Q

What are the cellular mechanisms of joint damage in RA?

A
  1. ACPAs enter the joints and bind citrullinated self epitopes in the cartilage/synovium
  2. Ab-Ag complexes bind Fc receptors and TLRS and activate inflammatory cells
    − Bind to macrophages → TNFa → major hallmark of disease in the joint
    − Can bind neutrophils → NETosis → releases cellular contents. This could include citrullinated peptides, providing a further source of antigen
  3. Complement is activated
  4. Synovial inflammation results
  5. Collagen and proteoglycans are degraded, eg) by MMPs (upregulated in chondrocytes in response to inflammation) → generates neoepitopes
  6. Neoepitopes taken up by DCs and presented to T cells – initiates viscious cycle of cellular resposnes
  7. Unresolved inflammation gives pannus formation, neo-vascularistiaon and lymphocyte accumulation
  8. Activation of chrondrocytes and differentiation/activation of osteoclasts as a result of the inflammatory process leads to progressive cartilage destruction and bone erosion
42
Q

What is the role of TNFa in RA?

A
  • Proinflammatory cytokine release → IL1, IL6, GM-CSF
  • Chemokine release → causes leukocyte accumulation
  • Hepcidin induction → acute phase response
  • PGE2 production
  • Osteoclast activation → bone resorption
  • Chondrocyte activation → MMP production and cartilage destruction
  • Angiogenesis
  • Endothelial cell activation → upregulation of E-selectin and VCAM-1
43
Q

What is the role of synovial fibroblasts in RA?

A

• Initial activation of SFs is due to synovitis:
– inflammatory cytokines - TNF, IL-1 and IL-6
– TLR ligands (e.g. RNA from nectrotic cells and matrix fragments) binding to TLR2, 3, 4.
• Activated SFs detectable early in RA contribute to initiation and perpetuation of disease by:
– Production of cytokines and chemokines - amplifies inflammation and inflammatory cell influx.
– Production of MMPs - causes cartilage breakdown.
– Production of VEGF and angiopoietin – promotes angiogenesis.
• SFs develop their permanently activated, aggressive phenotype (reduced apoptosis and enhanced proliferation) due to:
– Epigenetic factors, e.g. hyperacetylation and hypomethylation of DNA; modulate gene expression
– miRNA expression profiles influence the production of inflammatory cytokines etc.

44
Q

Describe pannus formation in RA

A
  • A pannus is an abnormal layer of fibrovascular tissue
  • In RA, the synovial lining expands and inflammatory cells are recruited
  • The expanded lining (the pannus) has invasive and descrutive characteristics
  • SFs produce RANKL – binds to RANK on osteoclasts and promotes activation of osteoclasts
45
Q

Describe the process and effects of cartilage degradation in RA

A

• Cartilage has load-bearing properties
• The pannus is a major cause of cartilage damage
− Protein binding characteristics of the cartilage change
− Invasion by SFs promoted
− Increased MMP synthesis
− Collagen network disassembled
− GAG and water content altered
• The cartilage has limited capacity for repair
− Avascular and depends on resident chondrocytes for its maintenance
− Chondrocyte apoptosis results in loss of maintenance
• Cartilage destruction results in joint space narrowing
• Normal protective effects of the synovium are lost

46
Q

Describe bone erosion in RA

A
  • Occurs rapidly, affecting 80% of patients within 1 year after diagnosis
  • RANKL expressed on activated T cells and SF binds to RANK on osteoclasts and activates them
  • Activated osteoclasts attach to bone and release degrading enzymes, eg) Cathepsin K
  • This process is actually essential to bone homeostasis, but in RA it is dysregulated due to elevated RANK::OPG ratios (OPG is a decoy receptor for RANKL), so bone erosion is excessive
  • Bone repair is inhibited due to cytokine-induced inhibition of osteoblast formation

ACPAs and osteoclastogenesis:
• ACPA with affinity for cVim bind osteoclast precursor cells and stimulate the rlease of TNF
• This enhances osteoclast activation

Evolution of bone erosion in the course of RA:
• ACPA produced during pre-clinical RA can stimulate osteoclast differentiation, leads to initial bone loss eg) in the marrow adjacent to the joint
• Synovitis leads to production of cytokines, which stimulate osteoclastogenesis by inducing expression of RANKL → enhances bone erosion and disease progresses.

47
Q

What is the treatment rational for RA?

A
  • Aim to achieve remission and prevent joint damage
  • Requires suppression of inflammation in the early stags of disease before irreversible joint damage
  • Improved tests and imaging methods allow earlier diagnosis
  • New treatments aim to target specific molecules/pathways that contribute to RA pathogenesis
  • Need to fully understand mechanism of early-stage pre-clinical RA to enable development of targeted treatments
  • Responses to treatments have been varied
  • Need for careful monitoring, so if it seems a patient is unresponsive, they can be switched to a new treatment quick enough
  • Biomarkers of disease progression would further improve clinical outcome

Treatment targets:
• In RA, inflammatory mechanisms are dysregulated. The absence of resolution leads to chronic inflammation
• Most treatments are therefore aimed at limiting inflammation
• Traditional treatments are NSAIDs and corticosteroids – reduce the effects of prostaglandins and relieve pain, but don’t affect disease progression.

48
Q

Describe anti-TNF as a DMARD for RA

A

DMARDs:
• Aimed at modifying the disease process by suppressing the immune response

Anti-TNF:
• The first biologic developed for the treatment of RA
• Cytokines including TNF and IL1 are overexpressed in the RA synovium
• Anti-TNF blocked production of IL-1, GM-CSF, IL-6 and IL-8 in synovial cell cultures → suggests TNF is the master regulator of inflammatory responses in RA
− Reduces the pro-inflammatory cytokine cascade
− Reduces leukocyte trafficking owing to decreased adhesion molecules and chemokines
− Reduced tissue destructive enymes
− Reduce angiogenesis
• In collagen-induced arthritic mice, anti-TNF inhibited inflammation, cartilage damage and bone loss
• Transgenic mice overexpressing human TNF develop RA-like disease
• Infliximab:
− Human/mouse chimeric TNF-specific mAb
− In combination with methotrexate (dihydrofolate reductase inhibitor commonly used to treat RA, inhibits activation of adhesion molecules in T cells) gave long lasting control of RA with inhibition of bone loss
− Infliximab + methotrexate now a common treatment for RA
− However, has to be given repeatedly – doesn’t cure the disease. Lak of sustained, drug-free remission may be due to suppression of some anti-inflammatory molecules as well as pro.

49
Q

What targeted treatments are apprived for RA?

A
Adalimumab
Etanercept
Infliximab	
-Cytokine inhibitor	
-TNFa	
-TNFa blockers were the first biological agents approved for the treatment of RA. TNF blockage has become a central strategy of targeted anti-inflammatory therapy in the disease.

Tocilizumab

  • Cytokine inhibitor
  • IL-6 receptor
  • This agent is considered the second major advance in cytokine blockade in RA. It has profound effects on systemic features, APR and synovitis.

Anakinra

  • Cytokine inhibitor
  • IL-1
  • Despite good anti-inflammatory activity in inflammasome driven disease, this agent has had only limited efficacy in RA

Rituximab

  • Cell-depleting agent
  • CD20 on the surface of B cells.
  • This is the only approved cell depleting agent for RA. Its use has reinforced the role of adaptive immunity, particularly humoral immune response, in the disease. Helps prevent production of anti-self antibodies.

Abatacept

  • Costimultion blocker
  • CD80 and CD86 – mimics activity of CTLA-4.
  • This agent disrupts the interaction of APCs with T cells, an effect that confirms the link between the innate and apative immune responses in RA. Will prevent the production of the inflammatory cytokines by APCs.
  • Work is also ongoing to see if we can inhibit the downstream signaling
  • Tofacitinib has been approved as a treatment of RA by inhibiting the JAK/STAT apthway
50
Q

Describe the role of pharmacogenetics in RA?

A

Pharmacogenetics
• RA progression and response to treatment is influenced by genotype
• Understanding this could lead to personalised treatments
• Responses to immunosuppresants, eg, methotrexate, can be altered by polymorphisms that affect drug transport, cytokine production and the folate pathway
• Up to 60% of patients don’t response well to anti-TNF treatments
− Responders have increased TNF, NFkB and HIF1a
− Non-responders have increased expression of genes involved in lipid and drug metabolism
• Biomarkers are needed to monitor responses
− Response to Rituximab can be monitored by measuring synovial B cell numbers
• GWAS highlited CDK6 and CDK4 as potential treatment targets for RA – these already have drugs against them for the cancer field, so may be possible to take drugs that are already well progressed in terms of testing, and use them for other indications

51
Q

What are the complications of RA?

A
  • RA doesn’t just affect the joints, it’s a systemic disease
  • Mechanisms that contribute to long term-complications:
  • Bone → low mineral density. Fractures (TNFa, RANK1)
  • Muscle → insulin resistance. (TNFa, IL1)
  • Fat → free fatty acid. Adipocytokines (TNFa, IL-6)
  • Liver → acute phase response. Iron redistribution (IL-6)
  • Blood vessels → Atherogenesis. Myocardial infarction. (Complement, IL-6)
  • Brain → low stress tolerance. Depression (IL-1, IL-6, TNFa)
52
Q

How are the systemic complications of RA treated?

A

Therefore, treatments should consider what is happening systemically and not just the joint
• RA is an independent risk factor for CVD
− Prevalence of atherosclerosis increased in RA patients
− Due to elevated cholesterol, systemic inflammation and oxidative stress
− Statins and ACE inhibitors can reduce RA inflammation and CVD risk
− Use of COX-2 inhibitors as anti-inflammatories in RA has been limited due to CV risks.

• RA is an independent risk factor for osteoporosis
− Increased risk of fragility fractures due to inflammation induced bone loss
− TNF, IL1, IL-17 and IL6 contribute to osteoclast stimulation
− Denosumab, mimics OPG and blcoks RANL binding to RANK, can inhibit bone loss

53
Q

What is meant by ‘tipping points’ in terms of RA?

A

A widespread misconception is that every step of the immune process is a potential target
• Not the case
• Most therapeutics only have a partial inhibitory effect, so only those molecules that are in short supply and are rate-limiting are likely to be useful targets
• Therapy that specifically targets most of the steps (so isn’t rate-limiting) yields little benefit in late disease
• The particular key molecules (eg, TNF) and the processes they control can be referred to as tipping points
− In epidemiology, a tipping point is the moment when epidemics reach a critical mass and have major repercussions
− This concept is valuable in autoimmune disease, as many cellular and molecule processes contribute to tipping the balance towards disease state, and therefore are potential targets.
− But although targeting these tipping points may provide benefit, blocking these critical physiological molecules may negative their benefitical roles in protective immunity

54
Q

Describe surgery as a treatment for RA

A
  • IN early phases of the disease, an arthroscopic or open synovectomy may be performed
  • Removes the inflamed synovia and prevents quick destruction of affected joints
  • Severely affected joints may require joint replacement surgery, such as knee replacement
  • Post-operative physio is always necessary
55
Q

What rodent models of RA are used?

A

Induced models of RA → injected with specific antigen.
• Synovial joints are the primary target of the autoimmune response in RA
• Immune responses (T cell and antibody) to cartilage proteoglycans (PG; e.g. aggrecan) have been detected in RA joints.
• In genetically susceptible strains of mice, immunisation with PG or collagen + adjuvant induces progressive arthritis (acute followed by chronic inflammation) that affects multiple joints.
• Mice with PGIA or CIA have:
– arthritic changes in the joints
– antigen-specific T and B cell responses
– elevated levels of inflammatory cytokines

Spontaneous models of RA:
• Knock-out or transgenic mice that spontaneously develop RA-like disease can be used to investigate the roles of specific gene products.
• Models include:
– human TNF-transgenic mice – develops chronic inflammatory erosive polyarthritis that is completely prevented by treatment with anti-TNF mAb.
– mouse transgenic for human HLA-DR0401, CD4, the auto-antigenic protein HCgp-39 and a TCR specific for a HCgp-39 epitope – has allowed investigation of Th1 responses in the context of HLA-DR0401
– K/BxN mice and serum-transfer-induced arthritis (STIA) – develop severe destructive inflammatory arthritis.

Development of the K/BxN mouse:
• TCR transgenic mice with specificity for a bovine peptide (RNase 42-56) restricted to I-AK MHC class II were crossed TCR-a chain-deficient mice with I-Ag7 class II alotype.
• By chance the transgenic TCR recognised a self peptide presented by Ag7 and the mice spontaneously developed arthritis due to the generation of autoreactive T and B cells.
• Arthritis in K/BxN mice is B and T cell dependent.
• Transfer of sera to recipient mice of different strains causes transfer of disease via antibodies
• The antibody specificity is for Glucose-6-phosphate isomerase (GPI)
• Antibodies to citrullinated GPI have been detected in a few cases of RA in humans

This is an example of molecular mimicry. T cells on the transgenic mice have receptors that can reocgnise a combination of the I-AK MHC class II with bovine RNAase, and can also recognize I-Ag7 class II molecule with GPI protein. Is an example of a T cell molecule that can recognize two different MHC:peptide complexes – this could be an example of what we see in human RA that enables it to recognise self peptide:MHC.

56
Q

How have mouse models provided evidence for mucosal mechanisms in RA?

A

• K/BxN mice are healthy if kept in completely germ-free environments
– Gavaging with segmented filamentous bacteria in the diet causes autoimmunity and inflammatory arthritis via a Th17-mediated response in the intestine and the periphery.
• Microbiomes of arthritis-susceptible DRB*0401 transgenic mice are dominated by clostridia; arthritis-resistant *0402 mice have Bifidobacteria and Porphyromonadaceae in their guts.
– *0401 mice have increased permeability of the gut mucosa and altered gene expression profile in their Th17 cells.
– suggests that the microbiome is influenced by genotype

57
Q

Describe what happened with the attempt to make anti-CD28 a treatment for RA

A

• TGN1412 (superMAB) = Anti-CD28. Mouse monoclonal antibody
• In mice → triggered Treg proliferation – possibly induced tolerance?
• Proposed for the treatment of RA and MS.
• In monkeys → no toxic effects
• First into human phase 1 trial began March 13, 2006
− Induced a cytokine storm (rather than inducing Tregs, induced CD4 effector memory cells), swelling, pain, vomiting, multi-organ failure, loss of consciousness.
− Controlled with steroid anti-inflamatory drugs and anti-IL2R antagonist.

58
Q

Describe the potential therapeutic effects of TSG protein

A
  • Working on this here at Manchester
  • TSG-6 expressed in RA joints in response to TNF and is protective in models
  • In CIA, TSG-6 transgenic mice have delayed onset of symptoms and reduced joint inflammation

How does TSG-6 act?
• TSG-6 inhibits neutrophil migration
− Extensive cartilage damage and bone erosion in TSG-6 KO mouse is accompanied by more rapid and extensive infiltration of neutrophils into joints
− In the murine air pouch model (mimics the articular synovium) → inflammatory stimuli injected into the pouch 15 mins after TSG-6 injected – neutrophil migration reduced by 40%
− Susbequently shown that TSG-6 inhibits neutrophil migration by binding to CXCL8
• TSG-6 inhibits expression of cartilage degrading enzymes by chondrocytes
− Both the full length protein and the link module fragment have an effect
− Very significant inhibition of protease → MMP-13 most significant
• TSG-6 inhibits RANK-L induced bone erosion
• TSG-6 inhibits osteoclast activity