Immunology Flashcards
what are the skin barriers to infection?
- tightly packed keratinised cells
- physiological factors: low pH, low oxygen tension
- sebaceous glands: hydrophobic oils repel, lysozyme destroys cell wall, ammonia/defensins = anti-bac
what are the mucosal surface barriers to infection?
- secreted mucous: physical barrier, secretory IgA (prevent entry), lysozyme, lactoferrin starves bacteria of iron
- cilia: trap/remove pathogens
how do commensal bacteria provide a barrier to infection?
- compete for resources
- produce fatty acids and bactericidins to inhibit growth
what are the cells of the innate immune system?
- polymohonuclear cells (neutrophils, eosinophils, basophils)
- monocytes, macrophages
- NK cells
- dendritic cells
what are the soluble components of the innate immune system?
- complement
- acute phase proteins
- cytokines and chemokines
what are the features of the cells of the innate immune system?
- identical in all
- express receptors to detect and home to sites of infection
- express PRRs to detect PAMPs/DAMPs at site of infection
- phagocytic capacity
- secrete cytokines/chemokines to regulate immune response
what are the different polymorphonuclear cells and where are they produced?
- neutrophils, eosinophils, basophils/mast cells
- produced in bone marrow, migrate rapidly to injury site
what do polymorphonuclear cells do?
- express receptors for cytokines = detect inflammation
- express PRRs = detect pathogens
- express Fc receptors for Ig = detect immune complexes
- phagocytosis
- oxidative and non-oxidative killing (esp neutrophils)
- release enzymes, histamine, lipid mediators of inflammation
- secrete cytokines = regulate inflammation
what are the types of mononuclear cells and where do you find them?
- monocytes, macrophages, lymphocytes
- produced in bone marrow
- circulate in blood
- migrate to tissues to differentiate into macrophages
what do mononuclear cells do?
- express receptors for cytokines = detect inflammation
- express PRRs = detect pathogens
- express Fc receptors for Ig = detect immune complexes
- phagocytosis
- oxidative/ non-oxidative killing
- secrete cytokines/chemokines = regulate inflammation
- APCs to T cells
how are phagocytes recruited?
- cellular damage/ bacterial products = local production of cytokines/chemokines
- cytokines = activate vascular endothelium = enhanced vascular permeability
- chemokines = attract phagocytes
how are microorganisms recognised?
- PRRs recognise PAMPs (bacterial sugars, DNA, RNA)
- Fc receptors bind to Fc portion of Ig
describe the process of opsonisation and endocytosis?
- endocytosis facilitated by opsonisation
- opsonins act as bridge between pathogen and phagocyte receptors
e. g. antibodies to Fc receptors, complement to complement receptors
what are the microbial killing mechanisms?
- oxidative
- non-oxidative
what is the process of oxidative killing?
- HOCl (hydrochlorus acid) acts as oxidant and anti-microbial
1. NADPH oxidase: oxygen to oxygen radical
2. superoxide dismustase: oxygen radical to hydrogen peroxide
3. myeloperoxidase: hydrogen peroxide with chloride to HOCl
what is the process of non-oxidative killing?
- release of lysozyme and lactoferrin into phagolysosome
- enzymes present in granules, can cover against bacteria and fungi
how are neutrophils involved in pus formation?
- phagocytosis depletes neutrophils glycogen reserves
- followed by neutrophil death
- as cell dies, enzymes released
- liquify local tissues
- accumulation of dying neutrophils = pus formation
- lots of pus = abscess
what is the aim of opsonisation?
to facilitate phagocytosis
what is the purpose of NK cells?
- present within blood
- migrate to inflamed tissues
- kills “altered self” or virus infected
- express inhibitory receptors for self-HLA molecules = prevent mal-activation by normal-self
- express activator receptors
- integrate signals from inhibitory and activator receptors
- secrete cytokines to regulate inflammation = promote DC function
where do dendritic cells reside and what do they represent?
- reside in peripheral tissue
- represent INNATE-ADAPTIVE transition
what do dendritic cells express?
- cytokine/ chemokine receptors = detect inflammation
- PRRs = detect pathogens
- Fc receptors for Ig = detect immune complexes
what are the 3 main functions of DCs?
- Phagocytosis (following this, DCs mature): upregulate expression of HLA, express co-stim molecules, migrate to lymph nodes
- present processed antigen to T cells in lymph nodes, prime adaptive immune response
- Express cytokines to regulate immune response
what are the components of adaptive immune system?
- Humoral immunity: B lymphocytes, antibodies
- Cellular immunity: T lymphocytes
- Soluble components: cytokines, chemokines
what are the characteristics of cells of adaptive immune system?
- wide repertoire of antigen receptors
- exquisite specificity
- clonal expansion
- immunological memory
what are primary lymphoid organs?
organs involved in lymphocyte development
- Bone marrow: T and B cells derived, B cells mature
- Thymus: T cells mature
what are secondary lymphoid organs?
sites of interaction between naïve cells and microorganisms
- spleen
- lymph nodes
- MALT
describe the process of T lymphocyte maturation
- arise from haematopoetic stem cells
- exported as immature cells to thymus where undergo selection
- mature T lymphocytes enter circulation, reside in secondary lymphoid organs
describe the selection and central tolerance of T cells
- low affinity for HLA = not selected (avoid inadequate reactivity)
- immediate affinity for HLA = positive selection ( 10%)
- high affinity for HLA = negative selection (avoid autoreactivity)
difference between the peptides recognised by CD8 and CD4 cells?
CD8+ T cells recognise peptide presented by HLA class 1 molecules CD4+ T cells recognise peptide presented by HLA class 2 molecules
what cells have MHC I vs MHC II?
- every cell has MHC I (HLA A, B, C)
- APCs also have MHC II (HLA DP, DQ, DR)
what do CD4+ helper cells do?
- recognise peptides presented on HLA class II molecules (APCs)
- immunoregulatory functions via cell interactions and cytokines (help develop full B cell response and some CD8+ T cell responses)
what do CD8+ cytotoxic killer cells do?
- specialised cytotoxic cells that kill cells directly (peforin, granzymes, Fas ligand)
- recognise intracellular peptides from HLA I
- secrete cytokines (IFNg, TNFa)
- defence against virus and tumours
why is T cell memory important?
response to successive exposures to antigen is bigger and stronger
act quicker
what are the different CD4+ T cell subsets?
- Th1
- Th17
- Treg
- TFh
- Th2
what do Th1 cells do? what cytokines involved?
Help CD8 T cells and macrophages
IL2, IFNg, TNFa, IL10
what do Th17 cells do? what cytokines?
help neutrophil recruitment, enhance generation of autoantibodies
IL17, IL21, IL22
what do Treg cells do? what cytokines?
IL-10/TGF beta expressing
express CD25 and Foxp3
what do TFh cells do? What cytokines in effector profile?
promote germinal centre reactions and differentiation of B cells into IgA and IgG secreting plasma cells
IL2, IL10, IL21
how does central tolerance of B cells work?
- no recognition of self in bone marrow = survive
- recognition of self in bone marrow = negative selection, avoid autoreactivity
describe the process of B cell activation
- B cell receptor binds to antigen
- some mature to plasma cells secreting IgM
- if signalled by CD4+ T cells in secondary lymphoid tissue, stimulated B cells rapidly proliferate
- undergo complex genetic rearrangement
what genetic rearrangements occur?
- isotype switching to IgG, IgA, IgE( TFh cells) - needs CD40:CD40L (not present in Hyper-IgM)
- somatic hypermutation: generate high affinity receptors
what are immunoglobulins?
soluble proteins made up of 2 heavy and 2 light chains
heavy chain determines class (IgM, IgG, IgA, IgE, IgD)
what are the functions of antibodys?
- identify pathogens/toxins (Fab-mediated)
- interact with other components of immune response (Fc-mediated): complement, phagocytes, NK cells
what are the features of B cell memory?
- lag time between antigen exposure to production is dec
- number of antibodies greatly increased
- dominated by IgG antibodies of high affinity
- independent of help from CD4+ T lymphocytes
what is important to remember about IgM secreting plasma cells?
generated rapidly following antigen recognition
not dependent on CD4 T cell help
summarise the process of B cells following antigen exposure
- Dendritic cells prime CD4+ T cells
- CD4+T cells help for B cell differentiation (requires CD40L:CD40)
- B cell proliferation, somatic hypermutation, isotype switching
what are complement?
> 20 types of proteins
produced in liver
present as inactive molecules in circulation
when triggered, enzymatically activate each other in biological cascade
what are the 3 ways of complement activation?
- classical pathway
- mannose binding lectin
- alternative pathway
describe the classical pathway
antibody + C1 –> C2, C4 –> C3
- changed antibody site exposes C1 binding site
- C1 binding to antibody activates cascade
- antibody-antigen immune complexes
describe the mannose binding lectin
MBL –> C2, C4 –> C3
- binding of MBL to microbial cell surface CHO
- direct stimulation of classical pathway (only C4, C2)
- not dependent on acquired immune response
describe the alternative pathway
PAMP (LPS, teichoic acid) –> C3
- C3 binds to bacterial cell wall components
- involved factors B, I, P
- not dependent on acquired immune response
what is the major amplification step in complement cascade?
C3
what effects does C3 activation have?
- inc vascular permeability and cell movement
- opsonisation of immune complexes = soluble
- opsonisation of pathogens = phagocytosis
- active phagocytes
- promote mast cell and basophil degranulation
- form MAC (via C5-C9) to punch holes in membrane
what is the function of cytokines?
- protein messengers
- immunomodulatory functions
- autocrine or paracrine dependent actions
what is the function of chemokines?
- direct recruitment/ homing of leukocytes in inflammatory reaction
- CCL19 and CCL21 are ligands for CCR7 and important in directing dendritic cells to lymph nodes
what is immunopathology?
damage to host caused by immune response
what is the basis of auto-inflammatory or auto-immunity?
immunopathology in absence of infection
what is the difference between auto-inflammatory and autoimmunity?
- autoinflammatory: driven by abnormalities in innate immune system (inappropriate activation of innate immune cells = damage)
- autoimmunity: abnormalities in adaptive immune system (abberant T and B cell responses in primary/secondary lymphoid organs, breaking of tolerance)
what factors can affect protein expression?
- DNA sequence
- genetics (mutations in DNA affecting protein sequence)
- epigenetics (heritable changes in gene expression e.g. via DNA methylation)
- microRNA (small non-coding ssRNA which target mRNA)
what other way can you sub-classify auto-inflammatory/ auto-immune diseases?
monogenic or polygenic
monogenic = rarer
what is the MOA of monogenic auto-inflammatory diseases?
- mutations in single gene encoding protein involved in innate immune cell function
- abnormal signalling via cytokine pathways (TNFa, IL1 common)
explain the inflammasome complex
- Toxins, microbial pathogens, urate stimulate cryopyrin
- Cryopyrin upregulates expression of ASC
- Pyrin-marenostrin decreases expression of ASC
- Apoptosis associated speck like protein (ASC) causes release of procaspase 1
- Procaspase 1 causes release of IL1, NFkappaB, apoptosis
- NFkappaB regulates TNFa
name a monogenic auto-inflammatory disease
Familial Mediterranean Fever
what is the pathogenesis of Familial Mediterranean Fever?
- AR
- mutations in MEFV gene = inactivated pyrin-marenostrin (expressed mostly neutrophils)
- get less downregulation of ASC
- so get increased pro-caspase 1 and increased inflammation driven by neutrophils
clinical presentation of FMF
- periodic fevers lasting 48-96 hours
- abdo pain (peritonitis)
- chest pain (pleurisy, pericarditis)
- arthritis
- rash
what is a long term risk of FMF and why?
AA amyloidosis
- liver produced serum amyloid A as acute phase protein
- deposits in kidneys, liver, spleen
- deposit in kidney = proteinuria, renal failure
what are other associations of AA amyloidosis?
- Auto-immune diseases: RA, ankylosing spondylitis, IBD
- auto-inflammatory diseases: FMF, muckle-wells syndrome
- chronic infections: TB, bronchiectasis, chronic osteomyleitis
- cancer: Hodgkin’s lymphoma, RCC
- chronic foreign body reaction
- HIV
what is associated with AL amyloidosis?
multiple myeloma
treatment of FMF
- colchicine (binds to tubulin in neutrophils = disrupt function)
- IL1 and TNFa inhibitors
what are the monogenic autoimmune diseases to know?
- abnormalities in tolerance = APS-1/ APECED
- abnormalities of regulatory T cells = IPEX
- abnormalities of lymphocyte apoptosis = ALPS
what is the pathogenesis of APS1/APECED?
- AR
- defect in autoimmune regulator AIRE (TF involved in T cell tolerance in thymus)
- upregulated expression of self antigens by thymic cells
- promotes T cell apoptosis
- defects in AIRE = failure of central tolerance = autoreactive T and B cells
what is the pathogenesis of IPEX?
- mutations in FOXP3 which is required for Treg development
- failure to negatively regulate T cell responses = auto-reactive B cells = autoantibody formation
what autoimmune diseases are involved in IPEX?
“diarrhoea, dermatitis, diabetes”
- enteropathy
- DM
- dermatitis
- hypothyroidism
what is the pathogenesis of ALPS?
- mutations in FAS pathway (cell surface death receptor)
- defect in apoptosis of lymphocytes
- failure of tolerance, failure of lymphocyte homeostasis
S/S of ALPS
- lymphocyte (lots)
- large spleen
- lymph nodes
- lymphoma
how do polygenic autoinflammatory diseases occur?
mutations in genes encoding proteins involved in innate immune cell function
characteristics of polygenic autoinflammatory diseases
- local factors at predisposed sites lead to activation of innate immune cells = tissue damage
- HLA associations less strong
- no autoantibodies
what genetic mutations have been found to be associated with Chrons (autoinflammatory)?
- NOD2 gene mutations (IB1 gene on Chr16)
where is NOD2 expressed and what is its role?
- expressed in cytoplasm of myeloid cells (macrophages, neutrophils, DC)
- is a cytoplasmic microbial sensor
- activation leads to TNFa release (inflammation) and it induces autophagy in DC
- abnormal degradation of DC = inflammation
treatment of Chrons
- corticosteroid
- anti-TNFa antibody
explain the polygenic nature of Chrons
- genetic mutations affecting innate immune response, epigenetic factors, microRNAs
- intestinal microbiota
- environmental factors e.g. smoking
these all come together to: - inc expression of pro-inflammatory cytokines/chemokines
- leukocyte recruitment
- release of proteases/free radicals
what is the MOA of mixed pattern diseases?
- mutations in genes encoding proteins in pathways of innate and adaptive immune cell function
- HLA associations may be present
- autoantibodies not usual
what is ankylosing spondylitis?
- genetic polymorphisms in >90%
- inflammation at specific sites of high tensile forces (e.g. entheses)
presentation of AS
- low back pain/stiffness
- enthesitis
- large joint arthritis
Tx of AS
- NSAIDs
- immunosuppression: anti TNFa or anti IL17
genetic polymorphisms of AS
- HLA B27: presents antigen to CD8 T cells
- IL23R: IL23 receptor and promoted differentiation of Th17 cells
- ILR2: inhibits IL1 so reduces TNFa (misfunction = inc TNFa)
features of polygenic autoimmune diseases
- mutations in genes encoding proteins in adaptive immune cell function
- HLA associations common
- autoantibodies found
why are HLA associations common?
HLA presentation of antigen is needed for development of aberrant T cell and T cell dependent B cell responses
break of tolerance, development of immune reactivity towards self antigens
what genetic polymorphisms are involved?
normally suppress T-cell activation but mutation means overactive T cells
- PTPN22: normally suppresses T cell activation
- CTLA3: expressed by T cells, transmits inhibitory signal to control T cell activation
in which diseases are allelic variants of PTPN22 found?
- SLE
- T1DM
- Rheumatoid arthritis
in which diseases are allelic variants of CTLA4 found?
- SLE
- AI thyroid disease
- T1DM
what is a type 1 hypersensitivity reaction?
- rapid allergic reaction
- pre-existing IgE antibodies to allergen
- IgE bound to Fc receptors on mast cell and basophils
- cell degranulation
- release of inflammatory mediators
what are the different inflammatory mediators that are released?
- preformed: histamine, serotonin, proteases
- synthesised: leukotrienes, prostaglandins, bradykinin, cytokines
what occurs as a result of these being released?
- increased vascular permeability
- leuckocyte chemotaxis
- SM contraction
what antigens usually cause a type 1 reaction?
FOREIGN
pollens, drugs, food, insect, animal hair
what is a type 2 hypersensitivity reaction?
Antibody reacts will cellular antigen
2 mechanisms of T2 disease = destruction OR activation/blockage]
how does antibody dependent destruction occur?
- complement activation –> cell lysis
- antibody binds and cause release of cytotoxic granules/ membrane attack from NK cells
- antibody binds enabling phagocytosis
name some syndromes that cause a T2 reaction and what the auto-antigen is against
- Goodpasture disease: non-collagenous domain of BM collagen type 4
- Pemphigus Vulgaris: epidermal cadherin
- Graves disease: TSH receptor
- Myasthenia gravis: ACh receptor
what is a type 3 hypersensitivity reaction?
- antibody reacting with soluble antigen to form immune complex
- immune complexes deposit in blood vessels
- inflammation and damage to vessels
what happens when immune complexes deposit in blood vessels?
- complement activation, infiltration of macrophages and neutrophils
- cytokine and chemokine expression
- granule release from neutrophils
- inc vascular permeability
name 2 syndromes that cause a T3 hypersensitivity reaction and what the autoantigen is
- SLE: DNA, histones, RNP
- RA: Fc region of IgG
what is a type 4 hypersensitivity reaction?
- delayed type
- T cell mediated response
how do you get tissue destruction in type 4 reactions?
- HLA I present self antigens to CD8 T cells = cell lysis
- HLA II present self antigens to CD4 T cells = cytokine production = inflammation and tissue damage
T cell recognises it as self = destroys it
what syndromes cause a type 4 reaction and what is the autoantigen?
- insulin dependent diabetes mellitus: pancreatic beta cell antigen
- rheumatoid arthritis: unknown synovial antigen
- MS/ experimental AI encephalitis: myelin basic protein
describe how T cells cause a T4 reaction?
- T helper cells activated by APC (presenting self antigen)
- when antigen it presented again in future, memory Th1 cells activate macrophages
- inflammatory reaction and tissue damage
what specific antibodies are seen in organ specific vs systemic diseases?
- organ specific diseases = organ specific antibodies
- systemic diseases = anti-nuclear antibodies
name some organ specific autoimmune diseases and the organ specific antibody
- Goodpasture disease = anti-BM antibodies
- Myasthenia gravis = anti-ACh antibody
- Graves’ diseases = anti-TSH receptor antibody
- Pernicious anaemia = anti-IF antibody
- DM = anti-GAD antibody
- Hasimoto’s thyroiditis = anti-thyroglobulin antibody
how is Graves disease mediated?
- IgG antibodies stimulate TSH receptor
- induce uncontrolled overproduction of thyroid hormones
- -ve feedback override antibody stimulation
what type of reaction is Graves?
type 2
how is Hashimoto’s thyroiditis mediated?
- associated with anti- TPO antibodies
- thyroid damage and lymphocyte inflammation
- goitre: enlarged thyroid infiltrated by T and B cells
what type of reaction is Hashimoto’s?
T2 and T4
how is type 1 DM mediated?
- CD8 T cell infiltration of pancreas = destruction of pancreatic islet cells
- T4 mediated
what antibodies are associated with T1DM?
antibodies pre-date development of disease
- anti-islet cell
- anti-insulin cell
- anti-GAD
- anti-IA-2
how is pernicious anaemia mediated?
- autoantibodies against IF
- no absorption of Vit B12
= pernicious anaemia and Sub-acute Combined Degeneration of Cord (SCDC)
what type of reaction is pernicious anaemia?
T2
how is myasthenia gravis mediated?
- autoantibodues against ACh receptors
- failure of depolarisation
- no muscle action potential
- T2 mediated
how can MG be diagnosed?
- Anti-ACh-R ABs
- EMG studies normal
- Tensilon test positive
how would you diagnose goodpastures?
- anti-BM antibody
- crescentic nephritis on biopsy
- T2 reaction
what are the genetic predispositions of rheumatoid arthritis?
- HLA-DR4 and HLA DR1 alleles: bind to citrullinated peptides with higher affinity
- PAD2 and PAD4 polymorphisms: associated with higher levels of citrullination
- PTPN22 polymorphism (+ CTLA4): involved in T cell activation
what are the roles of citrullinated peptides?
initiate inflammatory response
what environmental factors affect development of RA?
- smoking: erosive disease, increased citrullination
- gum infection with porphyromonas gingivalis (bacteria expressed PAD enzyme = promotes citrullination)
antibodies in RhA
- Anti-CCP antibody (more specific)
- Rheumatoid factor antibody
how are B cells involved in Rheumatoid arthritis?
T2 response: antibody binding to citrullinated proteins –> complement activation, macrophage activation, NK cell activation
T3 response: immune complex formation –> RF and anti-CCP, deposition with complement activation
how are T cells involved in Rheumatoid arthritis?
- T4 response
- APCs –> CD4+ T cell –> production of IFNy and IL17 –> increased IL1, TNFa
what are organ non-specific autoimmune diseases?
- characterised by presence of anti-nuclear antibodies
- very common
- test by staining of Hep2 cells
name some non-specific autoimmune diseased
- SLE
- systemic sclerosis
- idiopathic inflammatory myopathy
- ANCA-associated vasculitides
what are the symptoms of SLE?
- CNS: seizures
- Skin: butterfly rash, discoid lupus
- Heart: endocarditis, myocarditis, serositis, pleuritis, pericarditis
- Glomerulonephritis
- Haematological: HA, leukopenia, thrombocytopenia
- Arthritis and lymphadenopathy
genetic predispositions in SLE
- abnormalities in clearance of apoptotic cells
- abnormalities in cellular activation –> B cell hyperactivity and loss of tolerance
= antibodies directed particularly at intracellular proteins
what type of reaction is SLE?
T3 hypersensitivity
- antibodies bind to antigens to form immune complexes
- immune complexes deposit in tissue (skin, joints, kidney)
- immune complexes activate complement = classical pathway
- immune complexes stimulate cells expressing Fc and complement receptors
what is an important immunological investigation in SLE?
ANA
- measured by titre (minimal dilution at which antibody can be detected)
- “lumpy bumpy” on ELISA
targets of ANA
- anti-dsDNA AB (specific for SLE, high titre = severe disease, disease monitoring use)
- anti-ENA (extractable nuclear antigen) AB (Ro, La, Sm, U1RNP)
- anti-topoisomerase AB (diffuse CREST)
- anti-centromere AB (limited CREST)
how do these targets appear on ELISA?
- anti-dsDNA Ab = homogenous staining on ELISA
- anti-N Ab = speckled pattern on ELISA
which anti-ENAs are found in Sjogrens?
anti-Ro and anti-Lo
what other investigation can you do in SLE?
- measure complement
- formation of antibody-antigen immune complexes activates complement cascade via classical pathway
- depletion of C4 occurs first and then depletion of C3
- complement components become depleted if constantly consumed = act as marker of disease activity
what would the C3/C4 levels be in inactive, moderate and very active lupus?
- inactive lupus: normal C4, C4
- moderate active lupus: low C4
- very active lupus: low C3 and C4
symptoms of anti-phospholipid syndrome
- recurrent venous or arterial thrombosis
- recurrent miscarriage
- may occur alone (primary) or in with autoimmune disease (secondary)
what antibodies do you test for in anti-phospholipid syndrome?
- anti-cardiolipin antibody
- lupus anti-coagulant
types of systemic sclerosis
- diffuse cutaneous systemic sclerosis
- limited cutaneous systemic sclerosis
symptoms of diffuse cutaneous systemic sclerosis
- CREST features
- GI, pulmonary and renal involvement
- skin involvement of hands and PROXIMAL PAST forearms (unlike CREST)
ways of detecting DCSS
- anti-scl70 AB
- nucleolar pattern of immunofluorescence
symptoms of Limited cutaneous systemic sclerosis (CREST):
- only hands involved (does not spread proximally up forearms)
- involves peri-oral skin
Calcinosis Raynaud's Oesophageal dysmotility Sclerodactyly Telangiectasia
primary pulmonary HTN
Abs notes in CREST
anti-centromere AB
types of idiopathic inflammatory myopathy
- dermatomyositis (periorbital rash + Gottron’s papules = red/violet bumps on joints)
- polymyositis (no rash)
describe the mechanism of dermatomyositis
- within muscle = perivascular CD4 T and B cells
- immune complex mediated vasculitis
- T3 response
describe the mechanism of action of polymyositis
- within muscle = CD8 T cells surround HLA calss 1 expressing myofibers
- CD8 cells kill myofibers via perforin/granzyme
- T4 response
Ix in idiopathic inflammatory myopathy
POSITIVE ANA
- anti-Jo1 antibodies
- anti-Mi2
- Anti-SRP
what are the large vessel systemic vasculitis diseases?
- Takayasu’s arteritis
- GCA/polymyalgia rheumatica
what are the medium vessel systemic vasculitis diseases?
- polyarteritis nodosa
- Kawasaki disease
what are the small vessel ANCA-associated systemic vasculitis diseases?
- microscopic polyangiitis
- granulomatosis with polyangiitis (Wegener’s = crushed nose)
- eosinophilic granulomatosis with polyangiitis (Churg-Strauss)
what are the small vessel immune complex systemic vasculitis diseases?
- anti-GBM disease
- IgA disease
- cryoglobulinemia
what is ANCA?
- antibodies for antigens located in primary granules within cytoplasm of neutrophils
- inflammation = expression of these antigens on cell surface of neutrophils
- antibody engagement with cell surface antigens = neutrophil activation (T2 reaction)
- activated neutrophils interact with endothelial cells
- damage to vessels = VASCULITIS
what is cANCA?
- cytoplasmic fluorescence
- associated with antibodies to enzyme proteinase 3
- occurs in >90% of patients with GPA with renal involvement
what is pANCA?
- perinuclear staining pattern
- associated with antibodies to myeloperoxidase
- less sensitive/specific than cANCA
- associated with MPA and eGPA
what is the main difference between ANA and ANCA?
ANA = screening test for connective tissue disease ANCA = associated with subset of small vessel vasculitis, including MPA, GPA, eGPA
what type of virus is HIV-1?
retrovirus
genes = RNA molecules
in general how does HIV-1 replicate?
- inside a cell using reverse transcriptase to convert RNA to DNA to be integrated into host genome
what is the structure of HIV-1 virus?
- diploid genome
- contains 9 genes that encode 15 structural, regulatory and auxiliary proteins
what is the target of HIV-1?
CD4+ T helper cells, CD4+ monocytes, CD4+ dendritic cells
how does HIV affect CD4+ cells?
- uses host CD4 cells to replicate and move from cell to cell
- changes function of these cells
- so can’t mount full immune response
- lose immune memory = depleted memory T cells
- selective loss of CD4+ T helper cells
what does protection from HIV require?
- ABs ( B cells): prevent infection, neutralise virus
- sufficient CD8+ T cells to eliminate latently infected cells
what is the HIV-1 receptor and co-receptor?
- receptor: CD4 molecule/antigen
- co-receptor: CCR5 or CXCR4
what are the routes of transmission of HIV?
- sexually
- infected blood
- vertical
what is the process of natural immunity to HIV?
mobilised within hours of infection
- inflammation
- activation of macrophages, NK cells, complement
- release of cytokines/chemokines (those made by NK cells can reduce infection of CD4 cells by HIV)
- stimulation of plasmacytoid dendritic cells by TLRs
what is the process of acquired immunity (antibody and B cells) to HIV?
neutralising antibodies are produced
- anti-gp120 and anti-gp41 important in protective immunity
- non-neutralising anti-p24 IgG produced
- HIV remains infectious even when coated with antibodies
what is the normal role of CD4_ T cells (Th cells)? the cells killed by HIV infection!
- recognise processed antigens (esp Gag p24 peptides) from MHC II molecules
- coordinate immune response
what is the normal role in CD8+ T cells?
- can suppress viral replication (can kill HIV-infected cells or other malignant cells)
- secrete cytokines/chemokines to prevent infection –> block ebtry to CD4_ T cells
- recognise processed antigens from MHC II
summarise the effects of HIV
- activated infected CD4 T helper cells die
- infected CD4 T cells disabled
- MO/DC not activated by CD4+ T cells so cannot prime CD8+
- CD8+ T and B cell responses diminished without help
- CD4+ T cell memory lost
- infected MO/DC killed by virus/CTL = defect in antigen presentation, memory not activated
why is HIV prone to accumulate many mutations?
- replication of genome dependent on 2 low fidelity steps where errors can occur
1. RT (RNA to DNA) - lacks proof reading mechanisms
2. transcription of DNA into RNA copies
what advantageous features do these mutations allow the virus to gain?
- escape from neutralising Abs
- escape from HIV-1 specific T cells
- resistance and escape from ARVs
what are the seven steps in the life cycle of HIV?
- attachment and entry
- reverse transcription and DNA synthesis
- integration
- viral transcription
- viral protein synthesis
- assembly of virus and release of virus
- maturation
what are the different therapy targets within life cycle of HIV?
- attachment (Attachment inhibitors)
- reverse transcription (RTI)
- transcription of DNA to vural RNA
- viral protein cleavage by proteases (PI)
- fusion (FI)
- integration of viral DNA into host (INI)
- integration of viral RNA to produce viral proteins
- assembly and budding of new HIV
what ending do integrase inhibitors have?
- gravir
what ending do protease inhibitors have?
-avir
what ending do NRTIs have?
-ines
what is the median time from HIV infection to development of AIDS
8-10 years
what predicts progression and what are the different progressors?
viral burden predicts disease progression
- Rapid progressors (10%): 2-3 years
- Long-term non-progressors (LTNP <5%): stable CD4 and no symptoms after 10 years
- Exposed-seronegatives (ESN): repeatedly exposed to HIV, do not seroconvert
- Elite controllers (EC): suppress viral replication
what are the host genetic factors that can lead to someone being a LTNP?
- HLA profile (slow progressor)
- MBL alleles
- Gc Vit-D binding factor alleles
- heterozygosity for 32-bp deletion in chemokine-r CCR5
- TNF c2 microsatelitte alleles
what are the host immune response factors seen in a LTNP? (factors that keep HIV at bay)
- effective CTL, HTL and humoral responses
- secretion of: CD8 antiviral factor, IL-16, secretion of chemokines that block HIV entry co-receptors CCR5 and CXCR4
- maintenance of functional lymphoid tissue architecture
what virologic factor can cause a LTNP mechanism?
infection with attenuated strains of HIV
what are the 3 ways HIV can be detected?
detection via:
- anti-HIV antibodies (ELISA) = screening test
- anti-HIV antibodies (Western Blot) = confirmation test
- Viral load (PCR)
how is the CD4 count monitored and what does it correlate with?
- monitored via flow cytometry
- onset of AIDS correlates with decrease in number
what are the 2 ways to measure ART resistance?
- Phenotypic: viral replication measured in cell cultures under pressure of increasing concentrations of ARVs
- Genotypic: mutations detected by sequencing amplified viral HIV genome
what is the outcome of HAART?
- control of viral replication
- increase in CD4 T cell counts
- improvement in host defences
- does not eliminate HIV from body because there is reservoir in CD4+ T cells
what is the HAART regime?
combination of >=3 ART drugs
2 backbone drugs
1 binding agent
e.g. x2 NRTIs, x1 NNRTI or INI
what are the different types of backbone drugs?
- nucleoside reverse transcriptase inhibitors (NRTI) e.g. zidoviudine
- nucleotide RTI e.g. Tenofovir
- Non-NRTI e.g. Efavirenz
- Protease inhibitor e.g. Indinavir
what are the different binding agents?
- integrase inhibitor e.g. Raltegravir
- attachment inhibitors e.g. Maraviroc
- fusion inhibitors e.g. Enfuvirtide
what are the important HAART interactions to note?
- boosted PI: block cytochrome P450
- Efavirenz: block cytochrome P450
- INI: interacts with indigestion remedies
limitations and complications of HAART
- does not eradicate latent HIV
- threat of drug resistance
- high pill burden
- quality of life
- failure to restore HIV-specific T cell responses
- significant toxicities
- adherence
- cost
what are the different ways to prevent HIV spread?
- male circumcision (lots of APCs in foreskin)
- condoms
- PrEP (Truvada)
- Treatment as prevention (U=U)
what is an allergic disorder?
immunological process that results in immediate and reproducible symptoms after exposure to allergen
usually IgE mediated T1 reaction
what is an allergen?
harmless substance that can trigger an IgE mediated immune response –> clinical symptoms
what is sensitisation?
detection of specific IgE either by skin prick testing or in vitro blood tests
demonstrates risk of allergic disorder but doesn’t define it
what does Th1 and Th17 cells recognise?
- conserved structures in pathogens e.g. PAMPS
how are allergens differently detected and what cell leads the response?
- multicellular organisms and allergens don’t have conserved structures that are recognised by immune cells
- instead they release mediators (e.g. proteases)
- these disturb the epithelial barriers = functional change
- this is detected by immune system
- gives rise to Th-2 mediated responses
clarity the difference in the way that the Th1/Th17 immune response is initiated differs from Th2
- PAMP –> structural feature recognition –> Th1, Th17 immune response
- Helminths/allergens –> functional feature recognition –> Th2 immune response
explain the Th2 immune response
- allergens = stressed/damaged epithelium
- stressed epithelium release signalling cytokines (GM-CSF, IL-25, IL-33, TSLP)
- these act on Th9, Th2 and ILC2 cells and promote secretion of IL4, IL5, IL9 and IL13
- these act on eosinophils and basophils to expel parasites/allergens but also contribute to tissue injury
- TLSP can also act on Tfh2 cells to release IL4 and IL21
- The IL4 release from Tfh2 cells stimulated Bc ells to produce IgE and IgG4
describe the induction of Th2 immune response
- primary defect in epithelial barrier
- follicular dendritic cells (Langerhans cells and dermal DC) promote secretion of Th2 cytokines
- IL4 secretion is only induced by peptide-MHC presentation to TCR or naive/memory Th2 cells
how does the reaction differ between oral exposure and skin/respiratory exposure of allergens?
- oral exposure promotes immune tolerance
- skin/respiratory exposure induces IgE sensitisation
why does oral exposure promote tolerance?
- when allergen ingested through the oral route, T-regs (from GI mucosa) inhibit IgE synthesis
- oral tolerance requires induction of CD4+ T regs
- T regs inhibit multiple pro-allergic functions e.g. inhibiting DC APC function, secretion of IL-10 etc
describe the Th2 immune memory response when detecting an antigen
- mast cell is sensor and detects functional change
- allergen causes cross linking of IgE = histamine, prostaglandins, leukotrienes
- act on endothelium = inc permeability, SM contraction, neuronal itch
- expels parasite/allergen OR will be responsible for symptoms of asthma, eczema, hay fever
which cytokine plays a crucial role in development of Th2 immune responses? how is it induced?
IL4
only induced following peptide-MHC presentation to naive/memory Th2 cells
what causes the rapid and delayed symptoms of allergy?
- RAPID (2-3 hours): release of inflammatory mediators following allergen cross linking of IgE on surface of mast cells and basophils
- DELAYED: result of T2 cell (IL4, IL5, IL13) immune responses and eosinophil related tissue damage
- Th2 cytokines secreted by tissue lymphocytes act on effector cells to expel pathogens/allergens and repair tissue damage
why is the incidence of allergic disease increasing?
- hygiene hypothesis
- lack of Vit D in infancy (leading to food allergy)
- alternation of diversity in intestinal microbiome
- high conc of dietary advanced glycation end-products and pro-glycating sugars which immune system mistakenly recognises as causing tissue damage
how is allergic disease diagnosed?
- history
- examination
- allergen-specific IgE (sensitisation) test
- functional allergen test
what do you need to ask in the history of allergic diseases?
- age of onset of allergic diseasse
- occurs minutes to 3 hours after exposure
- symptoms (at least 2 organ systems involved)
- symptoms reproducible
- symptoms may be triggered by co-factors (i.e. NSAIDs in asthma)
allergic diseases that onset in infancy
- atopic dermatitis
- food allergy (milk, egg, nuts)
allergic diseases that onset in childhood
- asthma (house dust mite, pets)
- allergic rhinitis
allergic diseases that onset in adulthood
- drug allergy
- bee allergy
- oral allergy syndrome
- occupational allergy
what are the symptoms you should ask about?
- skin symptoms: urticaria, angioedema
- GI symptoms: D&V
- Resp tract symptoms: SOB, cough, wheeze
- vasculature symptoms: hypotension, impending doom
what is skin prick and intradermal testing?
- use skin test solutions, positive control (histamine) and a negative control (diluent)
- positive outcome = wheal >3mm than negative control
- antihistamines discontinued for 48 hours before
- more sensitive and specific than blood tests to diagnose allergy
advantages of skin prick tests
- rapid (read after 15-20mins)
- cheap/easy to do
- good NPV
- increasing size of wheals correlates with higher probability for allergy
- patient can see response
disadvantages of allergen-specific IgE tests
- requires experience to interpret
- risk of anaphylaxis
- high false positive rate
- can’t be used in patients with dermatographism/extensive eczema
what is the process of IgE RAST blood tests?
- allergen bound to sponge in plastic cup, patient’s serum is addedd
- specific IgE (is present) binds to allergen
- Anti-IgE antibody tagged with fluorescent label added
- amount of IgE/Anti-IgE measured by fluorescent light signal
what are the types of blood test allergens?
- aeroallergens: HDM, cat, dog, grass pollen mix, tree pollen mix
- food: peanut, tree nut, shellfish, egg, milk, what, soy
- venom: bee and wasp
- drugs: Penicillin G and V, ampilliin, insulin, tetanus
what are the indications of IgE RAST blood tests?
- pt who can’t stop antihistamines (otherwise do skin tests)
- pt with dermatographism
- pt with extensive eczema
- history of anaphylaxis
- borderline skin prick test results
what are the different functional allergen tests?
- in vitro tests: basophil activation, serial mast cell tryptase
- in vivo test: open or blinded allergen challenge `
what is the serial mast cell tryptase test?
- tryptase found in mast cell granules
- systemic degranulation of mast cells in anaphylaxis = inc tryptase
- peak concentration = 1-2 hours, baseline = 6-12 hours
- useful if diagnosis of anaphylaxis
what happens if serum tryptase doesn’t return to baseline?
? systemic mastocytosis
what is the basophil activation test?
- measurement of basophil response to allergen IgE cross-linking
- activated basophils inc expression of CD63, CD203, CD300 protein
- used in diagnosis of food and drug allergy
what is the open or blinded allergen challenge?
- GOLD STANDARD for food and drug allergy diagnosis
- inc volumes of offending food/drugs ingested
- under medical supervision
- risk of severe reaction
what is important to remember about allergen specific IgE sensitisation tests?
- used to detect the presence/absence of IgE antibody against external proteins
- positive IgE test demonstrates sensitization NOT clinical allergy
how can the serum IgE be used for prediction of allergic symptoms?
- concentration: higher levels = more symptoms
- affinity to target: higher affinity = increased risk
- capacity of IgE antibody to induce mast cell degranulation
what is component resolved diagnostics (CRD)?
- blood test to detect IgE to single protein components
- abundance and stability of protein contributes to risk of allergic disease
- useful for peanut and hazelnut allergy
what are the indications for allergen component testing?
- detect primary sensitisatoin
- confirm cross-reactivity
- define risk of serious reaction to stable allergens
- improve diagnosis for components poorly represented in whole food extracts
- improve diagnosis for components for unstable molecules in whole food extracts