Core Immunology Flashcards

1
Q

What is an allergy?

A

Undesirable, damaging, discomfort-producing and sometimes fatal reactions produced by the normal immune system directed against innocuous antigens in a pre-sensitized (immune) host.

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

What is type 1 hypersensitivity?

A
IgE
exogenous
15-30 minutes
wheal & flare
basophils & eosinophil
antibody
pollen-hay fever
allergic asthma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is type 2 hypersensitivity?

A

IgG/IgM Ab response against combined self/foreign antigen at the cell surface- complement activation/phagocytosis/ADCC

Clinical features
Onset minutes to hours
Cell lysis and necrosis

Common antigens: Penicillin

Associated diseases
Erythroblastosis fetalis,
Goodpasture’s nephritis

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

What is type 3 hypersensitivity?

A

IgG/IgM Ab against soluble antigen- immune complex deposition

Clinical features
Onset 3-8h

Vasculitis
Traditional cause-serum sickness
Associated diseases: SLE , arthritis, nephritis etc

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

What is type 4 hypersensitivity?

A

Antigen specific T-cell mediated cytotoxicity

Clinical features
Delayed onset 48-72h
Erythema induration

Common antigen
Metals-e.g nickel
(tuberculin reaction)
Associated diseases Contact dermatitis

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

Why do we get allergies?

A

Those components of the immune system implicated in allergic responses are primarily involved in responses to parasitic infection
The immune system has evolved to generate a rapid tissue-based response to re-infection
The lack of infectious drive is a contributory factor in allergic disease

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

What are the Immune responses to parasitic disease?

A
- Increased levels of IgE
   Total
    Specific to pathogen – cross-reactive
-Tissue inflammation with:
    Eosinophilia & Mastocytosis
     Basophil infiltration
-Presence of CD4+ T cells secreting:
    IL4, IL5 & IL13
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the Hygiene hypothesis?

A
  • Immune stimulation by microbes protects against allergies
  • Epidemiological data – Increase in Allergy
  • Animal Models – T1DM, EAE, Asthma
  • Increased atopy (Asthma) after anti-parasitic Rx
  • Prevention of autoimmunity (Crohn’s) by infections
  • Pro-biotics in pregnant women
  • Mechanism – Th1/Th2 deviation, antigenic competition, immune regulation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the Genetic influences on the ‘allergic’ immune response?

A
Polygenic diseases
Cytokine gene cluster IL3,5,9,13
IL12R; IL4R
FceRI
IFNg; TNF

NOT sufficient for disease
ONLY susceptibility

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

What are Susceptibility Genes for Allergic diseases?

A

Susceptibility genes for allergic disease.

Group 1: sensing the environment. The group of genes encodes molecules that directly modulate the effect of environmental risk factors for allergic disease. For example, genes such as TLR2, TLR4, and CD14, encoding components of the innate immune system, interact with levels of microbial exposure to alter the risk of allergic immune responses.71 Polymorphisms of glutathione-S-transferase genes have been shown to modulate the effect of exposures involving oxidant stress, such as tobacco smoke and air pollution on asthma susceptibility.

Group 2: barrier function. A high proportion of the novel genes identified for susceptibility to allergic disease through genome-wide linkage and association approaches have been shown to be expressed in the epithelium. This includes genes such as FLG,70 which directly affects dermal barrier function and is associated not only with increased risk of atopic dermatitis but also with increased atopic sensitization. Other susceptibility genes, such as ORMDL3/GSDML,34 PCDH1,24 and C11orf30,44 are also expressed in the epithelium and might have a role in possibly regulating epithelial barrier function.

Group 3: regulation of (atopic) inflammation. This group includes genes that regulate TH1/TH2 differentiation and effector function (eg, IL13, IL4RA, and STAT674; TBX21 [encoding T-box transcription factor]75; and GATA376), as well as genes such as IRAKM,77 PHF11,21 and UPAR23 that potentially regulate both atopic sensitization and the level inflammation that occurs at the end-organ location for allergic disease. This also includes the genes shown to regulate the level of blood eosinophilia (IL1RL1, IL33, MYB, and WDR36).41

Group 4: tissue response genes. This group includes genes that modulate the consequences of chronic inflammation (eg, airway remodeling), such as ADAM3317 and PDE4D,39 which are expressed in fibroblasts and smooth muscle, and COL29A1,25 encoding a novel collagen expressed in the skin linked to atopic dermatitis. Some genes can affect more than 1 disease component. For example, IL13 regulates both atopic sensitization through IgE isotype switching but also has direct effects on the airway epithelium and mesenchyme, promoting goblet cell metaplasia and fibroblast proliferation.14

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

What are allergens?

A

Antigens that initiate an IgE-mediated response

First encounter results in innate & IgM response

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

What is the Conventional Immune Response?

A

Allergen requires processing
Presentation to T cells & cytokine release
Results in delineation of T-helper subsets into different types

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

What is IgE?

A
  • IgE comes from B cells but helped by Th2 cells (which release IL-4)
  • IgE from plasma cells (differentiated B cells)
  • IgE is the main factor in allergic response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the Role of the Th2 T cell?

A
  • Generate Th2 type cytokines
  • Role of Th2 cell- recruits innate inflamm cells (cause tissue damage)
  • activate B cells to release IgE
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is Type I Allergic Response?

A
  • IgE binds to Fc recep on sensitised mast cell

* Allergen cross links IgE antigens- THIS is what causes allergic response

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

What is IgE Mediated Allergic Response?

A

Immunopathogenesis
• IgE Ab mediated mast cell and basophil degranulation- release of preformed and de novo synthesized inflammatory mediators
Clinical features
• Fast onset (15-30 min)
• Wheal and flare (immediate phase of this reac)
• Late phase response- delayed reacs
Late phase response
• Eosinophils
• Central role for Th2 T cell
• Late phase- from cytokines & phospholipid reacs

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

What is Anaphylaxis?

A
  • An acute, potentially life-threatening, IgE mediated systemic hypersensitivity reaction
  • Stage 1- sensitization (no allergic response at this stge- immune response exposed to allergen, T cells making allergic antibodies)
  • Stage 2- next time exposed- allergic antibosied made, IgE cross links
  • Late phase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What is the Atopic Triad?

A

Atopic Triad- Asthma, Rhinitis, Eczema
• Mast cells in airways linings cause symptoms
• Have immediate symptoms in asthma & delayed

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

What is Nasal Allergic Inflammation?

A
  • Allergic sensitization & allergic reaction in nasal mucosa= symptoms & functional alterations (e.g. nasal hyperresponsivness)
  • FcεR1 = high-affinity Fc receptor for IgE
  • GM-CSF = granulocyte-macrophage colony-stimulating factor
  • ICAM-1 = intercellular adhesion molecule-1
  • LFA-1 = lymphocyte function–associated antigen-1
  • MIP-1α = macrophage inflammatory protein-1α
  • NKA = neurokinin A
  • PAF = platelet-activating factor
  • TARC = thymus and activation-regulated chemokine
  • Treg = regulatory T cell
  • TxA2 = thromboxane A2
  • VCAM-1 = vascular cell adhesion molecule-1
  • VLA-4 = very late antigen-4
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is Rhinitis?

A

• Can be ALLERGIC/NON-ALLERGIC
• ALLERGIC= PERENNIAL (e.g. animals- exposed to throughout the yr House dust mite
Animal dander) or SEASONAL (hay fever related/ moulds)
Non-allergic: Vasomotor, Infective, Structural, Drugs, Hormonal, Polyps
• Blocked nose, runny nose - often with eye symptoms
• House dust mite, animal danders, pollens
• Treatment – Antihistamines & Nasal steroids (if don’t respond- can give immunotherapy)

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

What is Asthma?

A
  • Disease of INFLAMMATION & HYPER-REACTIVITY of small airways
  • In childhood - AERO-ALLERGIC stimuli - HOUSE DUST MITE key pathogenic importance
  • IMMEDIATE symptoms are IgE-mediated
  • DAMAGE TO AIRWAYS due to LATE PHASE RESPONSE (prostoglandins & leukotrienes)
  • DAMAGED AIRWAYS ARE HYPER-REACTIVE to non-allergic stimuli e.g. fumes (get symptoms from non-specific symptoms not just allergn e.g. perfume- not an allergn but hyperreactive airways )
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What is Atopic Dermatitis ?

A
  • DERMATITIS – MANY DIFFERENT TYPES
  • ATOPIC (inherited)
  • CONTACT - ALLERGIC/NON-ALLERGIC
  • CLINICALLY - Intense itching, blistering/weeping, cracking of skin
  • HOUSE DUST MITE now thought to be MAJOR TRIGGER in atopic disease
  • Topical Steroids & Moisturisers
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is the Interplay among the barrier, allergy, and pruritus ?

A

Interplay among the barrier, allergy, and pruritus as a trinity
• Itch- key component of pathogenesis
• Itch; disrupts barrier
• Cytokine can cause itch- treatments can target this

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

What is Sensitization and Acute Phase Response?

A
  1. Sensitization & Memory Induction
    • Sensitization to allergens & development of specific B-cell and T-cell memory
    • Allergen-specific T helper 2 (TH2) cells differentiation & clonal expansion= cytokine produc (interleukin-4 (IL-4) and IL-13)
    • Cytokines induce Ig class switching to IgE & clonal expansion of naive & IgE+ memory B-cell pops
    • IgE at surface of allergen-specific IgE+ B cells & other IgE-sensitized antigen-presenting cells facilitates antigen presentation.
    • T-cell activation in the presence of IL-4 increases differentiation into TH2 cells.
  2. Type 1 Hypersensitivity Reaction (IMMEDIATE phase of allergic reac)
    • Crosslinking of mast-cell & basophil cell-surface FcRI (high-affinity recep for IgE)-bound IgE by allergens
    • Leads to release of;
    o Vasoactive amines (e.g. histamine)
    o Lipid mediators (e.g. prostaglandin D)
    o Platelet-activating factor
    o Leukotriene C4 (LTC4), LTD4 & LTE4)
    o Chemokines (CXC-chemokine ligand 8 (CXCL8), CXCL10, CC-chemokine ligand 2 (CCL2), CCL4 & CCL5)
    o Other cytokines (such as IL-4, IL-5 and IL-13)
    • And to the immediate symptoms of allergic disease.
  3. Allergic Inflammation (LATE phase of allergic Reac)
    • Allergen specific T cells (under influence of chemokines & other cytokines) migrate to sites of allergen exposure, reactivated & clonally expand
    • Local IgE-facilitated antigen presentation by dendritic cells increases T-cell activation
    • Local IgE produc in allergic rhinitis & asthma but NOT in allergic skin inflamm (e.g. atopic dermatitis)
    • Eosinophils- one of main inflamm cells (up to 50% of cellular infiltrate) in asthmatic lungs but NOT in skin of those with atopic dermatitis
    • TH1 cells make interferon- (IFN) & tumour-necrosis factor (TNF)= contrib to activation & apoptosis of keratinocytes (in skin), bronchial epithelial cells & pulmonary smooth-muscle cells.
    Mast cells & basophils activation (release histamine, chemokines & other cytokines) also contrib to late-phase allergic reaction.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How do you diagnose allergies?

A
  • History
  • Specific IgE (>0.35 KuA/L)- TIME duration before symptoms
  • Skin prick test (>3mm wheal)- put drop of allergen extract on skin, scratch skin & will see weal & flare response
  • Intra-dermal test- (more invasive) inject allergen into intra-dermal space, see if size of bleb (mark you made when you have injected) increases
  • Oral challenge test – Gold standard- take 1/8th of a peanut & keep doubling dose- see if they can tolerate 4 or 5 peanuts (if can not allergic)
  • Basophil activation test- see if basophils change in surface proteins
  • Component resolved diagnostics
  • Immediate onset reacs- type 1 happens in few mins
  • Can measure specific antigens to common allergens- In vitro test (take blood sample form patient)
  • Take blood to see if antibodies against them- if are against parts of protin that are more stable- more severe reac
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What is the Basophil Activation Test?

A

Principle of the basophil activation assay;
• Upon cross-linking of membrane-bound IgE, basophils upregulate expression of specific activation markers e.g.CD63, CD203c & expression of the inhibitory receptor CD300a
• Exact mechanisms causing basophil degranulation remain elusive, but phosphorylation of p38 MAPK pivotal role in cell activation
• Put allergen on basophil

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

What are the Advantages and Disadvantages of the Specific IgE test?

A

Advantages: Safe
Disadvantages: False negatives, False positives

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

What are the Advantages and Disadvantages of the Skin Prick Test?

A

Advantages: Quick, Patient satisfaction
Disadvantages: False negatives, False positives, Antihistamines, Slight risk

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

How do you treat allergies?

A

• Symptomatic
o Antihistamines, Steroids, Adrenaline (if severe life threatening reac- patients can carry these if reac can affect airways)
o Specific – Immunotherapy (Subcutaneous or Sublingual)

• Indications
o Life threatening reactions to Wasp & Bee sting
o Severe Hay fever
o Animal dander allergy

• Not helpful
o Multiple allergies don’t respond to immunotherapy as well
o Food allergy
o Allergic rashes – Eczema, Urticaria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are the Mechanisms of Immunotherapy?

A
  • Allergen-specific immunotherapy (SIT) improves quality of life of treated individuals & reduces symptoms of allergy & medication use.
  • SIT; improved tolerance to allergen challenge, ↓ in immediate-phase and late-phase allergic inflamm, prevents development of new allergic sensitizations & ↓ progression of allergic rhinitis to asthma.
  • SIT is disease modifying & duration of effect beyond the treatment period.
  • SIT modifies cellular & humoral responses to allergen; ratio of T helper 1 (TH1) cytokines to TH2 cytokines increased & functional regulatory T cells induced.
  • Interleukin-10 (IL-10) produc by monocytes, macrophages, B cells & T cells is increased
  • The expression of transforming growth factor- (TGF) is increased & (with IL-10) TGF might contrib to regulatory T-cell function & immunoglobulin class switching to IgA, IgG1 and IgG4.
  • These immunoglobulins compete with IgE for allergen binding, decreasing allergen capture & presentation by IgE in complex with high-affinity recep for IgE (FcRI) or low-affinity recep for IgE (FcRII).
  • SIT also reduces mast cells and their ability to release mediators.
  • Eosinophils and neutrophils recruitment to sites of allergen exposure is also reduced
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are adverse reactions to food?

A

• Major Food Allergens (if person thinks allergic to something go through list on this slide);
o Water soluble glycoproteins 10 - 60 kd
o COW’S MILK (more common in childhood- 80% don’t end up with this allergy)
o EGG
o LEGUMES – PEANUT (if in childhood usully don’t always have this allergy when older) ; SOYBEAN; TREE NUTS
o FISH
o CRUSTACEANS / MOLLUSCS
o CEREAL GRAINS (refrain from testing to wheat as the wheat specific IG often +ve)
• Common- food intolerance rather than food allergy
• Pharmacological effect of food not allergic reac e.g. heart beats faster after coffeee
• Lactose intolerance- lack lactose recep
• Allergies; can get allergy syndrome (e.g. eat fresh apple- react, but don’t reac to cooked apple as proteins more sensitive, & when gets to stomach acid breaks protein down so don’t get reaction below airways)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What are clinical manifestations of adverse reactions to food?

A
  • Gastrointestinal: vomiting, diarrhoea, oral symptoms
  • Respiratory (upper & lower): rhinitis, bronchospasm
  • Cutaneous: urticaria, angioedema, role of food in atopic dermatitis unclear (rash for 2-3 wks unlikley is down to IgE sensitivity)
  • Anaphylaxis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What are drug allergies?

A
  • Cytotoxic- type II; hapten binds to protein on RBC- this combo recognised by immune system= haemolytic anaemia
  • If no response to eczema treatment creams- patch test them to see what allergic to
  • Chugg Strauss syndrome (serious)- don’t try testing them just tell them to avoid that drug
History
Indication for the drug
Detailed description of the reaction
Time between drug intake and onset of symptoms
Number of doses taken before onset

Management
Intradermal testing
Graded challenge
Desensitization

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What is the innate immune system?

A
  • Inflammation in target tissues (cells in tissues- respond to pathogens)
  • Cells; macrophages, dendritic cells, mast cells, neutrophils, complement
  • Pattern recognition against broad classes of antigen
  • No memory
  • No amplification
  • Little regulation
  • Fast response (hours – days)
  • Short duration
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

What is the adaptive immune system?

A
  • Cells circ through blood & coordinate responses
  • Cells; T & B cells
  • Highly specific (T and B cell receptors)- learn antigens that are foreign or not (memory)
  • Strong memory and amplification component (e.g. vaccines, previous infection)
  • Many regulatory mechanisms
  • Slow response (days to weeks for initial exposure)
  • Responses may last months - years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

How are the innate and adaptive immune system related?

A
  • Dendritic cells (innate) present antigen to T cells (adaptive)
  • T cell cytokines & B cell antibodies activate innate cells to cause inflammation
  • Innate immunity doesn’t adapt as well as adaptive immune system
  • Macrophage presents to T cell then get adaptive response
  • When T/b cells recognise foreign- they also active immune response (don’t work on their own)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are Phagocytic cells?

A
  • Neutrophils: eat and destroy pathogens (netosis- pus)
  • Macrophages: also produce chemokines to attract other immune cells
  • Dendritic cells (phagocytic): also present antigen to adaptive immune system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are Histamine-producing cells?

A
  • Mast cells, basophils, eosinophils: produce histamine and other chemokines and cytokines
  • Vasodilatation, attract other immune cells
  • Defence against parasites, wound healing but also allergy and anaphylaxis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What are Complement cells?

A
  • Directly attacks pathogens via alternative and lectin pathways
  • May be activated by adaptive immune system via antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What are Cytokines?

A

Signal between different immune cells (e.g. innate to adaptive, adaptive to innate)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are Chemokines?

A

Attract other immune cells to sites of inflammation (rather than signal)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is Autoimmunity?

A

• = the ADAPTIVE immune system (learned response to antigen) recognises and targets the body’s own molecules, cells and tissues (instead of infectious agents and malignant cells)
• Main characteristics:
o T & B cells that recognise self antigens
o B cells and plasma cells that make autoantibodies against self antigens
o Inflammation in target cells, tissues and organs is secondary to actions of T cells, B cells and autoantibodies
• Autoimmunity is more about the learning of what is self & non-self

Immunological disruption: Adaptive immunity
Main cellular involvement: B and T cells
Antibody involvement: Autoantibodies present
Clinical features: Continuous progression
Conceptual understanding: Breaking of self-tolerance
Main genetic susceptibility: MHC class II associations and adaptive response genes
Therapy: Anti-B and T cell
Examples: Monogenic ALPS and IPEX, Polygenic RA and
SLE

Autoimmunity
• Theoretical concept
• Many cells of the immune system have capacity for autoimmune functions
• Overlap with normal immune functions such as anti-tumour immunity
Autoimmune Disease
• Distinct clinical entities
• Breakdown of self-tolerance
• Environmental factors acting on favorable genetic background
• Wide variety of pathogenic mechanisms between diseases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is Autoinflammation?

A

• To distinguish from autoimmune diseases
• Main characteristics:
o seemingly spontaneous attacks of systemic inflammation (not learned T & B response)
o no demonstrable source of infection as precipitating cause
o absence of high-titre autoantibodies and antigen specific autoreactive T cells
o No evidence of auto-antigenic exposure

Immunological disruption: Innate Immunity
Main cellular involvement: Neutrophils, macrophages
Antibody involvement: Few or no autoantibodies
Clinical features: Recurrent, often seemingly unprovoked attacks
Conceptual understanding: Tissue-specific factors/danger signals
Main genetic susceptibility: Cytokine and bacterial sensing pathways
Therapy: Anti-cytokine (IL-1, TNF, IL-6)
Examples
Monogenic hereditary periodic fevers, polygenic Crohn’s disease, spondylarthropathies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are the 3 Factors Involved in autoimmune disease?

A
  • Genes can pre-dispose to certain disease e.g. certain MHC pre-disposes to rheumatoids but not just this as not born with disease
  • Env trigger acts on genetic background to cause disease
  • Immune regulation- certain regulation to help stop disease e.g. autoimmnity normally stops cancer (wipes out mutated cells before cause cancer) (regulation component can go wrong)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

How do you develop central tolerance of B and T cells?

A

• Have central & peripheral types of tolerance
• Central tolerance;
o B cells- Bone marrow
o T cells- Thymus
• T cells in thymus as develop- get a T cell recep (recognises antigen- has variability; can recognise anything e.g. pathogen or self antigen), then process of –ve selection (tested against self-antigens, if recep picks up antigen delete it), then +ve selection for useful T cells (e.g. if recep doesn’t recognise self- is +vly selected)
• T cell can be useful if only recognise at low level- regulatory
• B cells go through similar process in bone marrow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What are the functions of MHC1 and H@ and what genes encode them?

A
Class II MHC Genes May Shape T Cell Repertoire: Predisposing to Autoimmunity 
•	B cells & T cells variety to receptors they can express (not genetically linked) but can inherit types of molecules that leads to getting you autoimmune disease
•	MHC class I (on all nucleated cells in body- function is if there is something inside that tissue cells e.g. virus, gets presented on that surface) encoded by genes in HLA-A, HLA-B, HLA-C, On all nucleated cells, Presents antigen to CD8+ T cells
•	MHC class II (APCs e.g. skin infections, dendritic cell eats up antigen & takes it to lymph node to present to T cell in MHC) encoded by genes in HLA-DP, HLA-DQ, HLA-DR, On dedicated APCs, Presents antigen to CD4+ T cells
•	Have finite number of MHC types; each MHC subtype presents a certain antigen better than others e.g. if have a certain type of MHC might present self antigen better to T cells autoimmune (so usually have MHC class II association)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the consequences of a FOXP3 mutation?

A

FOXP3 mutation (gives regulatory cells)- failure to develop regulatory T cells – severe autoimmunity from birth

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are the consequences of a PTPNN22 mutation?

A

o PTPN22 mutations (genes that control how rapid activated T cells response is (how effective T cells are)- cause T cells to be activated more easily – stronger immune response in general

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What are the epidemiological factors of autoimmunity?

A

• Sex (hormonal influence)- women&raquo_space; men
• Age- autoimmunity more common in elderly (more exposed to diff env pathogen more likely to get autoimm disease)
• Sequestered Antigents
o May be recognised as foreign by the immune system (e.g. cell nucleus, eye, testis)
o Immune doesn’t react to certain cells as immune system never exposed to them so if happen to get exposed to them= autoimm
• Environmental triggers
o Infection
o Trauma-tissue damage
o smoking

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is Molecular mimicry ?

A

• Molecular mimicry e.g. in rheumatic fever antibodies against M protein of Streptococcus also happen to react against the glycoproteins of the heart valve cells (recep just happens to fit)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

How can changes to Autoantigens May Cause Autoimmunity ?

A
  • Citrullination of proteins make them more immunogenic (=rheumatoid arthritis)
  • Self antigens start to be recognised as foreign as antigen itself changed- a.a. citrulline not encoded into genome so will never make it in protein but can sometimes switch places with arginine (which was coded- what smoking does)- makes protein look a bit foreign to body
  • Tissue transglutamase alters gluten to help it bind to HLA-DQ (=coeliac disease)
  • Failure to clear apoptotic debris increases availability of sequestered antigens inside the cell (=systemic lupus erythamatosus)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

How do T cells stimulate autoimmunity?

A

• Th cell need antigen presented to them by APC with MHC
• T cell when sees antigen can;
o Differentiate into Th1 cell- make cytokines (signal to macrophages which make loads more cytokines)= inflammation
o Differentiate into Th2 cells= talk to B cells (cross talk)
• B cells differentiate into memory B cells & plasma cells= autoantibodies & cytokines=inflammation
• For process to stop; usually get rid of whatever causing it (e.g. infection)- but can’t do this in autoimmune disease (unless regulation)
Macrophages release inflammatory cytokines, this is also stimulated by T cells: APC binds to Th cell which causes Th1 to release inflammatory cytokines and Th2 to activate B cells causing auto antibody production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What is the Pathophysiology of autoimmune disorders?

A

• Autoreactive B cells and autoantibodies
o Directly cytotoxic
o Activation of complement
o Interfere with normal physiological function
• Autoreactive T cells
o Directly cytotoxic
o Inflammatory cytokine production
• General inflammation and end-organ damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

How do you group autoimmune disorders?

A

Organ Specific:
• Affect a single organ
• Autoimmunity restricted to autoantigens of that organ
• Overlap with other organ specific diseases
• Autoimmune thyroid disease is typical
Systemic
• Affect several organs simultaneously
• Autoimmunity associated with autoantigens found in most cells of body
• Overlap with other non-organ specific diseases
• Connective tissue diseases are typical

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What are General Clinical Features of autoimmunity?

A
  • More than 100 different diseases
  • Can affect any organ of body
  • Onset in middle age, old age
  • More common common in the elderly and women
  • Leads to loss of organ function
  • Lifelong-chronic condition
  • Characteristic exacerbation and remission
  • Traditionally divided into organ specific or systemic
  • Common for diseases to overlap
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is Autoimmune Thyroid Disease?

A

• Hashimotos thyroiditis (underactive thyroid)
o Destruction of thyroid follicles by autoimmune process
o Associated with autoantibodies (only in thyroid gland) to thyroglobulin & to thyroid peroxidase
o Leads to hypothyrodism
o 1st get hypEr as lots of thyroxine released but end up with hypo
o Inflamm & damage

• Grave’s disease
o Antibody mimics TSH- binds to TSH recep on thyroid galnd which stimulates recep which makes gland over active
o Inappropriate stimulation of thyroid gland by anti-TSH-autoantibody
o Leads to hyperthyroidism
o Graves- just antibody causes disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What is the cause of Myasthenia Gravis?

A
  • Acetylcholine usually stimulatess muscle contraction
  • Myasthenia gravis- antibody against ACh receptors these antibodies block receptors so no acetylcholine stimulation therefore patients have decreased muscle activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What is the cause of Pernicious Anaemia?

A

• Can’t absorb B12- as intrinsic factor can’t bind it as autoantibody blocks intrinsic factor (withoit binding to intrinsic factor Vit B12 cant be absorbed) , can get parathesia , fatigue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

What is Lupus?

A
  • Antinuclear antibodies against structures in cell nucleus=systemic autoimmune disease as cell nucleus everywhere in body
  • Reason for butterfly rash- when go out in sun, UV light causes cells to undergo apoptosis, breaks down & contents of cell on outside of cell (in bleb), so the nuclear antigen visible to immune system (no longer hidden in cell)
  • Non-organ specific- autoantibodies against antigens in the nuclus combine with their targets forming immune complexes in the circulation these can land anywhere in body (not necessarily where antigen came from e.g. skin) so can cause inflammation anywhere that’s why things like renal problems are a common side effect
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

What are Connective Tissue Diseases?

A
  • Systemic lupus erythematosus
  • Scleroderma
  • Polymyositis
  • Sjogrens syndrome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are the General Principles of Diagnostic Testing ?

A
  • Should use diagnostic tests to answer specific questions/ support clinical diagnosis NOT as screening tools
  • Need to have an idea of what your testing for before ordering diagnostic test
  • Their ability to correctly discriminate between health & disease is improved when they are used in the appropriate population
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What is Sensitivity & Specificity? How are the calculated?

A

A: test positive and disease positive
B: test positive and disease negative
C: test negative and disease positive
D: test negative and disease negative

  • Sensitivity [a/(a+c)]- True positive: how good the test is in identifying people WITH the disease (ability to pick up affected individuals)
  • Specificity [d/(c+d)]- True negative: meausure of how good the test is in correctly defining people WITHOUT the disease
  • Low specificity if test also picking up people without disease
  • Positive predictive value [a/(a+b)]-The proportion of people with a positive test who have the target disorder
  • Negative predictive value [d/(c+d)]- The proportion of people with a negative test who do not have the target disorder.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What are the different types of autoimmune diagnostic tests?

A
  • Non specific- Inflammatory markers

* Disease specific- Autoantibody testing, HLA typing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What are Non-Specific Markers of Systemic Inflammation?

A
  • ESR
  • CRP- acute phase response
  • Ferritin- acute phase response protein (high)
  • Fibrinogen- acute phase response protein
  • Haptoglobin
  • Albumin
  • Complement (e.g. serum C3 or 4- e.g. systemic autoimm disease- low complement= active episode)
  • Can get +ve blood test indicating lupus- doesn’t mean is active disease
  • Need other investigations to guide decision & assessment of patient
  • Inflamm markers & complementary tests- in someone to determine if active disease
65
Q

What are Antinuclear Antibodies (ANA)?

A

Antibodies in the patient’s blood that bind to the cell nucleus- can be then more specific and identify subtypes of antibody that bind to different bits of the cell nucleus

Sensitivity & Specificity of Specific Anti-Nuclear Antibodies
• Can find ANA in people that had just had illness, can find ANA in older people (aging immune system), or ANA in an autoimmune illness to come (but don’t mean anthing at that moment in time)
• So can’t just look at ANA on it’s own

66
Q

How do you detect ANA?

A
  • Detect autoantibodies; put cells of interest on microscope slide, use fibroblasts (large nuclei to see- will have more of antigens in them), then incubate slides with patient serum, , if have autoantibodies in patients serum auto-Abs will stick to antigens on cells, use antiIgG against Auto-Abs which are fluorescently labelled
  • Nuclei flouresence
  • If have autoantibodies in patients serum auto-Ab will stick to antigen on cells
  • Antihuman Igg
  • Pattern on nucleus (e.g. homogenous, speckled etc) depends on specific target antigen
67
Q

How do you detect dsDNA & ENA’s?

A
Techniques;
•	Anti-dsDNA
o	Crithidia luciliae assay (protosoa)
o	Farr assay
o	ELISA
•	ENA’s
o	Immunoblots
o	Individual ELISA’s
o	Combination of antigens
•	>100 different antibodies described in SLE
•	Look for more specific targets 
•	Smash up nucleus & smear nuclear material across paper, will see molecular mass of certain proteins that will be seen on block, use 2ndry antibody to detect
68
Q

What is Multiplex assessment of non‐organ‐specific autoantibodies with a novel microbead‐based immunoassay?

A
  • Attach beads to antigens of interest (know what targets are)- beads have flouresence that you can detect using lasers
  • If have something attached to bead 2ndry antibody seen in diff colour
  • Incubate serum with beads, if have mulitple autoantibodies attach to their specific beads
  • Need to interpret info in clinical context
  • Disadv- looking for normal autoantigens (occasionally patients with +ve test which is non-specific)
69
Q

What are Ro, La , CCP, Scl antibodies specific for?

A
Ro: neonatal/ Sjorens syndrome
dsDNA and Sm, RNP etc: SLE
La: Sjorens syndrome, SLE
CCP: Rheumatoid Athritis
Scleroderma: ANA positive for Scl 70 Antibody
70
Q

What is Rheumatoid Factor?

A
  • RF is an antibody (IgM, IgG or IgA) directed against the Fc portion of IgG
  • Commonly found in rheumatoid arthritis but NOT diagnostic of the diseases (sensitivity & specificity around 70%- as RF can come after infection, RF physiological role to mop up unnecessary antibodies in blood)
  • RF can be seen with other diseases in which polyclonal stim of B cells is seen (e.g. chronic infections)
  • High titters may be pathogenic in vasculitis (high levels of RF sticky protein- forms complexes- can precipitate out of circulation= stick to vessel wall=activate complement=vasculitis)
  • Common test for autoimmune condition
  • Can auto aggregate
71
Q

What is Anti-CCP ?

A
  • ACPA more specific (95%) for rheumatoid arthritis then Rheumatoid Factor
  • Similar sensitivity to RF but more specific
  • Useful prognostic marker
  • ACPA positive patients tend to have more severe and erosive disease
  • If patient has citrulated antibodies- more aggressive disease
72
Q

How do you diagnose Rheumatoid Arthritis?

A

 The diagnosis of Rheumatoid Arthritis is clinical
 Combination of symptoms, Xrays, blood tests
 High RhF, CCP and elevated ESR and CRP add to the probability of a definitive diagnosis
 Early treatment can prevent joint damage and significant morbidity.

73
Q

When do you order a Anti-neutrophilic Cytoplasmic Antibodies (ANCA) test? What is it?

A

Ordered when ?vaculitis- e.g. renal or ENT

• Incubate neutrophils with patients serum, use 2nsry autoantibodies to see what attached to neutrophils

ANCA is the pattern and anti-PR3/MPO is the autoantibody causing the pattern

Cytoplasmic (c)ANCA: Granular fluorescence of neutrophil cytoplasm with nuclear sparing. Target antigens: PR3 (90%) and MPO
Perinuclear (p)ANCA: Apparent fluorescence of the nucleus only: Target antigens: MPO (70%) and PR3

74
Q

What are the features of Anti-neutrophilic cytoplasmic antibodies (ANCA) associated systemic vasculitis?

A

Histology: Leukocytoklastic vasculitis; necrotising, granulomatous inflammation
Ear, Nose, Throat: Nasal septum perforation, saddle-nose deformity, conductive or sensorineural hearing loss,
subglottic stenosis
Eye: Orbital pseudotumours, scleritis, episcleritis, uveitis (50%)
Lung: Nodules, infiltrates, or cavitary lesions, alveoral haemorrhage
Kidney: Segmental necrotising glomerulonephritis
Heart: Occasional valvular lesion
Peripheral nerves: Vasculitic neuropathy (10%)
Eosinophilia : Mild eosinophilia occasionally

75
Q

What is the Clinical utility of ANCA testing?

A
  • How good it is to predict certain conditions
  • Can’t rely entirely on autoantibody for diagnosis (this is to support diagnosis)
  • Positive ANCA is extremely useful in suggesting the diagnosis in the proper clinical setting
  • Histopathology remains the gold standard for diagnosis in most cases (e.g. if renal or cardio involvement)
  • Negative ANCA assays do not exclude AASV (vasculitis) since 10%-50% of patients may be ANCA neg
  • Persistence of ANCA in the absence of clinical indications of active disease does not indicate a need for continued treatment (see how succesful treatment is)

• Reemergence of ANCA pos in a patient who was ANCA neg whilst in remission suggests a risk of disease flare. The temporal correlation between the return of ANCA and a disease flare is poor

WG: 80%-90% ANCA positivity and PR3»MPO 75%-90%:5%-20% ANCA Antigen specificity
MP: 70% ANCA positivity and PR3»MPO 50%-80%:10%-25% ANCA Antigen specificity
CSS: 50% ANCA positivity and PR3»MPO 2%-50%:3%-35% ANCA Antigen specificity

76
Q

What is Autoimmune liver disease?

A
  • Anti-mitochondrial Ab specific for primary biliary sclerosis
  • Anti-smooth muscle and anti-liver/kidney/microsomal (LKS) Abs, found in autoimmune hepatitis
  • These antibodies (above) can be present before disease presents
  • Antibodies detected by IF screening using rodent tissue block (oesophagus, liver and kidney) and antigen specific ELISA
  • Use tissue block ; bits of kidney, liver, stomach- depending on immunoflouresence get can predict specific autoantibodies relevant to autoimmune liver disease
77
Q

What Autoantibodies are in Type I DM

A
•	Non-pathogenic
•	Several types:											
o	islet cell antibodies								
o	anti-GAD65  anti-GAD67								
o	anti-insulinoma antigen 2 (IA-2)			
o	insulin autoantibodies (IAAs)
•	Disappear with progression of disease and total destruction of β islet cells 
•	Type 1 autoimm diabetes- autoimm antibodies against islet cells in pancreas=what sort of antibodies developed e.g. 1 type in circulation or can have wide spread (shows disease progressing- have established type 1 diabetes, so to try prevent coming to this point can try use markers to help put in place intervention) 

Role of Autoantibodies in Diagnosis of Type I DM
• Disease conformation
• to identify relatives and patients at risk of developing autoimmune diabetes
• Negative predictive value of ICA and IAA is almost 99%
• Increased risk of disease development with greater n.o. of diff autoantibodies present & younger age

78
Q

What is the Future of diagnostic testing in investigation of autoimmune diseases?

A
  • Cytokines determination in serum
  • Detection of antigen specific autoimmune T and B cells
  • T-reg detection, ? measure of therapeutic response
  • Personalised medicine, genetic profiling to determine individual risk of the disease and to tailor the most appropriate therapy
  • Discovering increasing number of biomarkers some more useful than others
79
Q

What is immunodeficiency? What are the two types?

A

Clinical situations where the immune system is not effective enough to protect the body against infection
• Primary- inherent defect in immune system (usually genetic) e.g. component may not be working, usually severe
• Secondary- immune system affected due to external causes e.g. HIV infec

80
Q

What are Secondary Causes of Immunodeficiency ?

A
  • Breakdown in physical barriers: CYSTIC FIBROSIS (defect in mucous in resp tract)
  • Protein loss (when loose proteins e.g. Igs can’t remake enough or can loose proteins e.g. by weeing them out (kidneys))- burns, protein loosing enteropathy, malnutrition
  • Malignancy- especially lymphoproliferative disease, myeloma (cancer of plasma cells- take up all the space in bone marrow so not enough space for other cells)
  • Drugs- steroids, DMARDS (disease modifying anti-rheumatic drugs e.g. methotrexate), Rituximb (mono-clonal antibody- used clinically to try to eradicate cells e.g. can use in rheumatoids), anti-convultants, myelosuppresisve (treat leaukaemias- kills good & bad cells so get immunodeficiency)
  • Infection- HIV, TB
81
Q

What are Phagocytes?

A
  • Neutrophils, macrophages
  • Eat bacteria/ fungi= destroys them
  • Bacterial infection= neutrophils (e.g. in chest infec green sputum- neutrophils, if white- tends to be a viral infec)
  • Macrophages in tissues (monocytes in blood)- start local inflamm response
  • In macrophage (lysosome & phagosome fuse)
82
Q

What are Pattern Recognition Receptors ?

A

• Pathogen recognition receptors (PRRs)- recognise conserved pathogen associated molecular patterns (PAMPs) (unique to each pathogen) e.g. lipopolysaccharide
• Phagocytes use PRRs to detect pathogens
• Toll like receptors- a type of PRR;
o TLR3- recognises viral RNA (defects in TLR3 lead to recurrent HSV encephalitis)
o TLR4- recognises lipopolysaccharide (on lots of bacteria)
o TLR5- recognises flagellin (makes up flagella on bacteria)
• Cascade of events when receptor triggered with intracellular molecules= production of inflammatory cytokines
• MyD88 & IRAK4 are involved in this cascade

83
Q

What are Toll like receptors?

A

a type of Pattern Recognition Receptor

84
Q

What does TLR3 recognise?

A

recognises viral RNA (defects in TLR3 lead to recurrent HSV encephalitis)

85
Q

What does TLR4 recognise?

A

recognises lipopolysaccharide (on lots of bacteria)

86
Q

What does TLR5 recognise?

A

recognises flagellin (makes up flagella on bacteria)

87
Q

What does activation of Toll like receptors result in?

A
  • Cascade of events when receptor triggered with intracellular molecules= production of inflammatory cytokines
  • MyD88 & IRAK4 are involved in this cascade
88
Q

What is IRAK4 deficiency?

A

IRAK4 DEFICIENCY
• Clinical presentation;
o Recurrent bacterial infection (especially strep & staph)- pneumonia, meningitis, arthritis
o Poor inflammatory response (person with pneumonia should have raised CRP but because cascade broken they don’t have raised CRP)
o Susceptibility to infection decreases with age
• Rx; prophylactic antibiotics, IV immunoglobulin if severe
MyD88 presents similarly

89
Q

What is a Phagolysosome ?

A
  • Inside phagocyte
  • NADPH complex- several proteins stuck together including gp91phox. This complex makes electrons which bind to O2- to form hypochlorus acid production
  • This kills bacteria
90
Q

What is the cause of Chronic Granulomatous Disease?

A
  • Granulomas- collection of macrophages
  • Defect in gp91phox
  • Macrophage sees bacteria but can’t do anything, so calls more macrophages but they can’t do anything- end up with collection of macrophages

Typical presentation
History of recurrent skin abscesses, 1 x previous pneumonia
Presented to hospital with a liver abscesses
Mothers brother-similar but milder symptoms
Investigations:
Normal immunoglobulins, lymphocytes and neutrophil count

91
Q

What is Chronic Granulomatous Disease?

A
  • Recurrent abscesses; lung, liver, bone, skin, gut
  • Unusual organisms; staphylococcus, Klebsiella, serretia, Aspergillus, Fungi
  • Rx; haemopoetic stem cell transplant, antibiotics
  • Tests rely on REDUCTION (gain of electron)
  • Get patient’s neutrophils & activate them- then measure how many neutrophils reduce dihydrorhodamine
  • Measure Dihydrorhodamine reduction using flow cytometry
  • Nitro blue tetrazolium (NBT) dye reduction- healthy neutrophils should go purple (NBT reduced)
92
Q

What is Complement?

A

Non immunoglobulin proteins, that are important in the immune system for
Cell lysis (kill invading bacterium)
Control of inflammation
Stimulate phagocytosis
Sequence of events activates complement proteins
• Classical pathway- bacteria (c1 antigen antibody= cascade)
• MBL pathway similar to classical pathway (but mannose binding lectin acitaves cascade)
• Sequence of events activates complement proteins
o C3a, C4a & C5a- arteriole dilation, histamine release form mast cells, chemotaxis of phagocytes
o C3b- microbe opsonisation promotes phagocytosis
o C5b, C6, C7, C8, C9- form MAC (causes microbe cytolysis- punches holes in it)
• C5b binds C6 & 7= C5b67 complexes bind to memb via C7= c8 binds to complex & inserts into cell memb of pathogen= C9 bind to complex & polymerise=1-16 molecules of C9 bind to form a pore in the memb

93
Q

What is the function of Complement?

A
  • Lyse foreign cells that are covered in antibody
  • Test; antigen RBC & antibodies against RBC (of sheep), drop on patients serum & if MAC working RBCs should lyse)- this test looks at all components to see if working
94
Q

What conditions often have decreased levels of C2-C4?

A

SLE, infections, myositis

95
Q

What does recurrent bacterial meningitis indicate?

A
  • C5-9 (forms memb attack complex)- repeated episodes of BACTERIAL meningitis (particularly Neisseria meningitis
  • if get more than one bacterial meningitis- THIS IS NOT NORMAL
96
Q

How does the binding of antibodies to antigens inactivate antigens?

A
  • Neutralisation (blocks viral binding sites/ coats bacteria)
  • Agglutination of bacteria
  • Percipitation of dissolved antigens
  • Or activation of complement
97
Q

What is x-linked agammaglobulinaemia?

A
  • Defect in Bruton’s Tyrosine Kinase (BTK)- need for B cell signalling & maturation
  • BTK DOWNSTREAM of B cell recep so if this blocked B cell can’t respond- this happens in the the bone marrow so B cells don’t mature downstream so may not have any mature B cells
  • B cell maturation NOT completed in bone marrow
98
Q

What is the presentation of x-linked agammaglobulinaemia?

A

Male presented before age of 5; 3 hospital pneumonia admissions, recurrent chest infecs in between, sinusitis. Mother’s brother passed away age 35 from bronchiectasis (suggetss x linked recessive).
• Bloods;
o No B cells, normal T cells
o No IgG, IgA, IgM
o CT: bronchial thickening, evidence of recent pneumonia

99
Q

What are common B cell defects? What do they cause/ how are they treated?

A
  • CVID (common variable immunodeficiency) – low IgG, IgM, IgA
  • IgA deficiency– is found in entry of mucosal surfaces
  • X-linked hyper IgM syndrome (1st antibody B cells if IgM then G (which is better) then A)- cell machinary can’t switch to making other Igs so have lots of M but noth much IgG or A
  • Transient hypogammaglobulinaemia of infancy (4-6mnths antibodies should usually be produced)
  • Loss of antibody secretion
  • Usually leads to recurrent bacterial infec with pyogenic organisms (bacteria that cause pus)
  • Treat with antibiotics then IV IgG for life
  • Most very serious
  • Some less serious e.g. IgA deficiency; 1 in 5-700, some completely well, higher risk of autoimmune disease e.g. coeliac
100
Q

What is a common cause of secondary antibody deficiency? What is the presentation? How would you treat this?

A

50 yr old female, rheumatoid arthritis history (severe), no infecs most of adult life but 2 yr history of recurrent bacterial chest infecs (5 courses of antibiotics in winter). PMH: asthma. Drug Hx: currently on methotrexate & infliximab, previously rounds on Rituximab. Has also had gold, sulfasalzine in past.
• Not likley to be genetic defect or would have had lots of infecs before too
• Ix (investigations): slightly low B & T cells, low IgG & IgA

Has secondary antibody deficiency due to drugs (comparatively common)- as drugs she’s on depress immune system
• Knocks out B cell so eventually also get lower plasma cells
• Others affect T cells

Treatment:
• Antibiotics
• Immunoglobulin G replacement (as IgG makes up most of blood, IgM lower & A usually in secretion)
• Can give it IV or subcutaneously

101
Q

What is Severe Combined Immunodeficiency (SCID)?

A

SCID (Severe Combined Immunodeficiency)
• Diagnosis: no T cells & suggestive history
• Investigations;
o Normal IgG levels, NO IgA, reduced IgM
o Lymphocyte makers show absent/ reduced T & NK cells but present B cells

• RX:
o Peadiatric emergency
o Antibiotics, antivirals, antifungals
o Asepsis
o Haemopioetic stem cell transplant (only cure)
o Screen

SCID casues;
o Defect/ absence of critical T cell molecule (TCR, coomon gamma chain)
o Loss of communication- MHCII deficiency (T cells can’t help B cells so pretty non functional)
o Metabolic- adenosine deaminase deficiency (also have bone deformities & neurological defects)

Presentation may be a baby with fulminant chicken mix and recurrent infections (often consanguineous parents)

Defects in T Cells
• (If pateint had no T cells- PAEDIATRIC EMERGENCY)
• Usually more severe as B cells also need T cell help (so even if tehre are B cells they don’t function)
• Symptoms; recurrent infec with opportunistic infections, bacteria, viruses
• Fungi (candida), protozoa (pneumocystis)
• Types;
o SCID
o Others e.g. DOCK8 deficiency (B & T cells can’t communicate)

102
Q

What is Immunomodulation?

A
  • Immunomodulation- Using immunomodulatory drugs to manipulate the immune system to get the desired immune response
  • A therapeutic effect of immunomodulation may lead to immunopotentiation, immunosuppression, or induction of immunological tolerance
103
Q

What are the Mechanisms of Immunomodulation?

A
  • Immunization
  • Replacement therapy
  • Immune stimulants or suppressants
  • Anti-inflammatory agents
  • Allergen immunotherapy (desentization)
  • Adoptive immunotherapy
104
Q

What are Biologics?

A

Definition
Medicinal products produced using molecular biology techniques including recombinant DNA technology

Main classes
Substances that are (nearly) identical to the body’s own key signaling proteins
Monoclonal antibodies- •Used in rheumatoids
• Anti-TNF drugs e.g. Infliximab (but has murine variable domain- own immune response against it so ineffective)
• Cetrolizumab- just has Fab region with chain for stability
Fusion proteins- Entercept

105
Q

What is Immunopotentiation?

A
  • Immunopotentiation- enhancement of immune response by increasing it’s speed, extent & duration
  • Immunization; passive or active
  • Replacement therapies
  • Immune stimulants
106
Q

What is Passive Immunisation?

A

• Definition: transfer of specific, high-titre antibody from donor to recipient. Provides immediate but transient protection
• Problems:
o Risk of transmission of viruses
o Serum sickness
• Types:
o Pooled specific human immunoglobulin
o Animal sera (antitoxins an antivenins)- animals injected with toxin, have antibody response
• Uses:
o Hep B prophylaxis and treatment
o Botulism, VZV (pregnancy), diphtheria, snake bites

107
Q

What is Active Immunisation?

A
•	Definition: to stim development of a protective immune response & immunological memory
•	Immunogenic material: 
o	Weakened forms of pathogens
o	Killed inactivated pathogens
o	Purified materials (proteins, DNA)
o	Adjuvants 
•	Problems: 
o	Allergy to any vaccine component
o	Limited usefulness in immunocompromised (either don’t respond to vaccine/ risk of having the infection)
o	Delay in achieving protection
108
Q

What are Replacement Therapies & Immune Stimulation ?

A
  • Pooled human immunoglobulin (IV or SC)- used in RX of antibody deficiency states
  • From blood donors; plasma fractionation, keep immunoglobulins
  • G-CSF/GM-CSF- act on bone marrow to increase production of mature neutrophils
  • IL-2 (Stimulates T cell activation- rarely used as IL-2 used in regulatory cells more so IL2 may be better used in autoimmune conditions)
  • α-interferon (Main use in treatment of Hep C)
  • β-interferon (Used in therapy of Multiple Sclerosis)
  • γ-interferon- certain intracell infecs e.g. salmonella (atypical mycobacteria), chronic granulomatous disease & IL-12 def
109
Q

What drugs cause immunosuppression?

A
  • Cortocosteroids
  • Cytotoxic/ agents
  • Anti-proliferative/activation agents
  • DMARD’s (disease modifying anti-rheumatic drugs)
  • Biologic DMARD’s
110
Q

What is the action of corticosteroids?

A
  • Can suppress innate & adaptive immune systems (not very specific effect)
  • Decreased neutrophil margination (so neutrophil count will go up)
  • Reduced production of inflammatory cytokines
  • Inhibition phospholipase A2 (reduced arachidonic acid metabolites production)
  • Lymphopenia (as toxic to T cells)
  • Decreased T cells proliferation
  • Reduced immunoglobulins production (e.g. if given steroid drugs)
111
Q

What is the side effects of corticosteroids?

A
  • Carbohydrate and lipid metabolism; diabetes, hyperlipidaemia
  • Reduced protein synthesis- poor wound healing
  • Osteoporosis
  • Glaucoma and cataracts
  • Psychiatric complications
112
Q

What are the uses of corticosteroids?

A
  • Autoimmune diseases- CTD (connective tissue disease), vasculitis, RA
  • Inflammatory diseases- Crohn’s, sarcoid, GCA/polymyalgia heumatic
  • Malignancies- Lymphoma
  • Allograft rejection
113
Q

What are targets for T Cell Targeted Immunosuppression ?

A
  • Normally APC engages T cell which recognizes antigen=T cell stim= makes IL2 cytokine=allows spread of T cell activation (progression of immune response) Tacrolimus stops this
  • Cytokines= transcription of genes
  • Prolif stage= M-TOR signalling molecule allows T cells to proliferate - Sirolimus inhibits M-TOR
  • Drugs prevent different stages (targeted)
  • Anti IL-2 R mAbs blocks IL2 which also stops T cell-T cell activation
  • Azathioprine blocks T cell proliferation
114
Q

What are classes of drugs that Target Lymphocytes ?

A
  • Antimetabolites; Azathioprine, Mycophenolate mofetil (MMF)
  • Calcineurin (serine/ threonine phosphatase) inhibitors; Ciclosporin A (CyA), Tacrolimus (FK506)
  • M-TOR inhibitors- Sirolimus
  • IL-2 receptor mABs- Basiliximab
115
Q

What is the function of Sirolimus?

A

Sirolimus (Rapamycin)
• Macrolide antibiotic
o Also binds to FKBP12 but different effects
o Inhibits mammalian target of rapamycin (mTOR)
• Binds mTOR- T cells can’t proliferate (stuck in G1-S phase)
• Mode of action- inhibits response to IL-2
• T cell effects- cell cycle arrest at G1-S phase

116
Q

What is the function of Calcineurin Inhibitors ?

A

• CyA- binds to intracellular protein cyclophilin
• Tacrolimus (FK506)- binds to intracellular protein FKBP-12
• Cyclophilin & tacrolimus- immunosupressants by preventing T cell activation
• Mode of action;
o Prevents activation of NFAT
o Factors which stimulate cytokines (i.e IL-2 and INFγ) gene transcription
• T cell effects- reversible inhibition of T-cell activation, proliferation and clonal expansion

117
Q

What are the side effects of Calcineurin/ mTOR drugs?

A
  • Hypertension
  • Hirsutism (abnormal hair growth on woman’s face & body)
  • Nephrotoxicity
  • Hepatotoxicity
  • 2ndry lymphomas (due to long term immunosuppression)
  • Opportunistic infections
  • Neurotoxicity
  • Multiple drug interactions (induce P450)
118
Q

When are Calcineurin/ mTOR dugs clinically used?

A
  • Transplantation- allograft rejection

* Autoimmune diseases

119
Q

What are Antimetabolites ?

A

Inhibit nucleotide (purine) synthesis (interfere with DNA synthesis- so cell can’t multiply as can’t synthesis enough DNA, so will affect organs that have lots of cell proliferation)
• AZA (azathioprine)
o Guanine anti-metabolite
o Rapidly converted into 6-mercaptopurine
• MMF
o Non-competitive inhibitor of IMPDH (inosine monophosphate dehydrogenase)
o Prevents production of guanosine triphosphate
• T and B cells effects
o Impaired DNA production
o Prevents early stages of activated cells proliferation

120
Q

What is the action of Methotrexate ?

A

Folate antagonists

121
Q

What is the action of Cyclophosphamide?

A

Cross-link DNA (so DNA synthesis will be impaired- cells won’t survive)

122
Q

What are the Side Effects of Cytotoxics?

A

All can cause:
o Bone marrow suppression & susceptibility to infections
o Gastric upset (nausea)
o Hepatitis

  • Cylophosphamide- cystatis (bladder inflamm)
  • MTX- pneumonitis
123
Q

What are the Clinical Uses of Cytotoxics?

A
  • AZA/MMF- autoimmune diseases (SLE, vasulitis, IBD), allograft rejection
  • MTX- RA, PsA, Polymyositis, vasculitis, GvHD in BMT
  • Cyclophosphamide- vasculitis (Wagner’s, CSS), SLE
124
Q

What are Biologic DMARD’s?

A
Biologic DMARD’s (can target specific molecules)
•	Anti-cytokines (TNF, IL-6 and IL-1)
•	Anti-B cell therapies
•	Anti-T cell activation
•	Anti-adhesion molecules 
•	Complement inhibitors

i.e : RA is treated with anti TNF tolicizumab, Anti-IL-17 mAb, Rituximab and Abatacept

125
Q

What is Anti - TNF drugs?

A

Anti-Cytokines
o First biologic drugs successfully used in therapy of RA (5 different agents now licensed)
o Used in other inflammatory conditions (Crohn’s, psoriasis, ankylosing spondylitis)
o Earlier use the drug- better the effect (before get further along in the pathogenesis)
o Caution: increase risk of TB

126
Q

What is Tocilizumab?

A

Anti-Cytokines - Anti-IL-6
o Blocks IL-6 receptor
o Used in therapy of RA and AOSD
o May cause problems with control of serum lipids

127
Q

What is Anti IL-1 drugs?

A

Anti-Cytokines
o 3 different agents available (anakinra, rilonacept and canakinumab)
o Used in treatment of AOSD and autoinflammatory syndromes

128
Q

What is Rituximab?

A

• Chimeric mAb targets CD20 on B cell surface
• First approved in ’97 for treatment of chemotherapy resistant DLCL
• Only CD20 molecule in memory & transitional B cells so won’t affect all B cells- early B cells survive so immune system can reform
• Many uses:
o Lymphomas, leukaemias
o Transplant rejection
o Autoimmune disorders

129
Q

What is Adoptive Immunotherapy? When is it used?

A
•	Bone marrow transplant (BMT)
•	Stem cell transplant (SCT)
•	Take progenitor cells out- kill all cells- put progenitor cell back 
•	Uses 
o	Immunodeficiencies (SCID)
o	Lymphomas and leukemias
o	Inherited metabolic disorders (osteopetrosis)
o	Autoimmune diseases
130
Q

Which Immunomodulators are used for Allergy?

A
  • Treatment of allergy usually steroids
  • Immune suppressants
  • Allergen specific immunotherapy- more targeted
  • Anti-IgE monoclonal therapy
  • Anti-IL-5 monoclonal treatment

Allergen Specific Immunotherapy
• Indications;
o Allergic rhinoconjunctivitis not controlled on maximum medical therapy
o Anaphylaxis to insect venom
• Mechanisms;
o Switching of immune response from Th2 (allergic) to Th1 (non-allergic)
o Development of T reg cells & tolerance
• Routes- SC or sublingual for aeroallergens
• Side effects localized & systemic allergic reactions

131
Q

What is Omalizumab?

A
  • mAb Targets IgE
  • Used in Rx of asthma (where conventional therapy failed)
  • Also useful in Rx of chronic urticaria and angioedema
  • May cause severe systemic anaphylaxis
132
Q

What is Mepolizumab?

A
  • mAb against IL-5
  • Prevents eosinophil recruitment and activation
  • Limited effect on asthma
  • No clinical efficacy in hypereosinophilic syndrome
133
Q

What are innate defences?

A
  • Skin (barrier, sebum, normal flora) – e.g. burns
  • Mucous membranes (tears (keep eye surface clean), urine flow, phagocytes)
  • Lungs (goblet cells, muco-ciliary escalator (bacteria back up to surface). Cystic fibrosis- mucus stays in lungs)
  • Interferons, complement, lysozyme, acute phase proteins (not produced in reaction to a specific threat but just available)
  • Normal commensal flora in gut – antibiotic treatment alters flora e.g. can get C. difficile, Candida spp (CLOSTRIDIUM) infections.
  • (Extremes of age, pregnancy, malnutrition- more prone to infection)
134
Q

When can oral thrush occur?

A

• Can occur after taking antibiotics

135
Q

What infections are burns patients at risk of?

A

At risk of;
• (Staph aureus)
• Pseudomonas infec
• Group A strep infecs

136
Q

What are neutrophils?

A
  • Neutrophils (NE) very important after initial breach of innate defences
  • Less – increasing infection
137
Q

What are qualitative/quantitative neutrophil defects?

A

• Qualitative defects (neutrophils don’t work properly) (e.g. lose ability to kill or chemotaxis) or
Chemotaxis – rare, congenital, e.g. inadequate signalling

• Quantitative defects (not enough neutrophils- less present) Killing power - inherited, e.g. Chronic Granulomatous Disease. At risk of Staph. aureus infections

138
Q

What is Neutropenia?

A

 Quantitative defects lack of neutrophil
 Causes;cancer treatment, bone marrow malignancy, aplastic anaemia - drugs
 “Neutropenic” – not enough neutrophils
 Esp. imp. <0.5x10^9/L (neutrophil count where starts getting bad) and if prolonged (more risk of infec)
 >50% proceed to infection. High mortality
 Empirical therapy - Concept
 >50% those with Pseudomonal infections will die in 24hrs if not treated
 Need to give antibiotics that have Pseudomonas cover e.g. piperacillin tazobactam (broad spec & covers pseudomonas)

139
Q

What can affect Neutropenic Patients?

A
  • Bacterial infections – e.g E. coli (in bowel- if ulcers get into blood), S. aureus (can get inside if break in skin)- often normal flora e.g. Coag neg staph (line infecs)
  • Fungal infections – Candida spp.(candidia in eye, or line infec) , Aspergillus spp. (spores inhaled- chest infecs)
  • Treatment – broad spectrum antibiotics.
  • e.g. Antipseudomonal penicillin +/- gentamicin (affects gram –ves), 2nd line treatment e.g. a carbapenem (meropenem)
  • fungi, viruses
  • Granulocyte stimulating factors (GCSF)- if neutropenic can induce GCSF to try to stim bone marrow making neutrophils again
140
Q

What are T cell deficiencies?

A
  • Congenital – rare – T helper dysfunction +/- hypogammaglobulinaemia
  • Acquired – drugs e.g. ciclosporin after transplantation (decreases graft versus host disease and rejection), steroids
  • Acquired – viruses e.g. HIV (affects T cells)
141
Q

What Opportunistic Pathogens are common in T cell deficiencies?

A

• May be intracellular
• BACTERIAL – Listeria monocytogenes (associated with soft cheese), Mycobacteria
• VIRAL – stem cell transplants - HSV (herpes simplex), CMV, VZV. Serological testing, prophylaxis and treatment with e.g. aciclovir and ganciclovir (antivirals)
• FUNGAL – e.g. Pneumocystitis spp., Candida spp., Cryptococcus spp. (cryptococcal meningitis marker disease of HIV)
(If someone has TB check for HIV too)

cell deficiencies – Protozoan and Parasitic infections
• Cryptosporidium parvum - oocysts shed by cattle/humans. Faecal oral route. Most patients recover after prolonged illness of up to 3 wks (if immunocomp. can go on longer). May take much longer in T-cell deficients. Symptomatic treatment only (in most cases)
• Toxoplasma gondii- in cat faeces (preg ladies a bit T cell defieicent)
• Strongyloides stercoralis- worm invades through skin, makes it’s way to gut (mostly asymptomatic but if immunosuppressed can get overgrowth of worms & hyperreaction, woems can go throughout body esp in lungs)

142
Q

What are Hypogammaglobulinaemias?

A
  • Antibody problems!!
  • Congenital – e.g. X-linked agammaglobulinaemia (rare)
  • Acquired – multiple myeloma, burns
  • Usually ENCAPSULATED bacteria that cause problems e.g. S. pneumoniae
  • Parasitic that cause problems- e.g. Giardia lamblia
  • Treatment – Immunoglobulin (to replace antibodies- BUT every time give transfusion risk of giving infection)
143
Q

What is Complement Deficiency?

A
  • Hereditary, rare
  • Encapsulated bacteria most important organisms. Need complement to help kill organisms.
  • e.g. C5-8 then Neisseria meningitidis (gram –ve diplococci) is important in this particular complement deficiency
  • Frequent, serious S. pneumoniae infections as poor quality opsonisation
144
Q

What is a Splenectomy? Why is it preformed?

A
  • Spleen - source of complement and antibody producing B-cells, removes opsonised bacteria from blood.
  • Causes - traumatic, surgical or functional e.g. sickle cell anaemia
  • Streptococcus pneumoniae, Haemophilus influenzae type B, N. meningitidis, malaria
  • High mortality, vaccination, prophylactic penicillin (if don’t have spleen to protect against the above microoganisms), education – seek help if unwell
145
Q

What are Biologics?

A
  • Are antibodies or other peptides- inhibit inflammatory cytokine signals e.g.tumour necrosis factor or TNF, inhibiting T-cell activation, or depleting B-cells.
  • E.g. used in severe rheumatoid arthritis
  • Risk of tuberculosis, herpes zoster, Legionella pneumophila, and Listeria monocytogenes
146
Q

What is the relationship between organ transplants and infections?

A

• Anti-rejection treatment suppresses cell mediated immunity to stop effects of cytotoxic T cells and natural killer cells. Degree of immunosuppression varies on how closely the donor and recipient are matched and organ involved.
Diary of infections would get in course of organ transplantation
• 1. The initial disease e.g. HBV
• 2. Surgery and hospital admission e.g. S. aureus wound infection
• 3. Organ receipt (organ they’re getting comes with risk) e.g. Toxoplasmosis, CMV
• 4. Opportunistic infection during initial immunosuppression (initial 3/12, e.g. CMV, Aspergillus)
• 5. Later on get opportunistic infection (after 3/12, e.g. Zoster, Listeria)- more T cell type infecs
candid albicans can occur from line insertions give

147
Q

Why can you have blood group incompatibility?

A
  • Immunofluorescent staining revealed blood group substances in cell membranes of all vascular endothelial cells & certain epithelial cells not just RBCs.
  • Normal individuals have naturally occurring anti-A or anti-B isoagglutinins (why you can have incompatibility)
  • Poor outcome of transplants performed between blood group incompatible individuals.
148
Q

What are MHC?

A
  • Set of genes found in all vertebrate species
  • Important role in immune function, disease susceptibility and reproductive success. ‘
  • Proteins encoded by the MHC are expressed at the cell surface and function to present ‘self’ and ‘nonself’ antigens for inspection by T cell antigen receptors.
  • 50,000 – 100,000 MHC molecules on the average mammalian cell (most common protein on cell surface)
  • MHC- mark your cell as yours so anything else as foreign
  • Highly polymorphic.
  • Role in histocompatibility, major influence on graft survival.

The Human MHC
• 6p21.3
• 3.6Mbp
• Also known as ‘the HLA complex’
• Divided into three regions, class I, II and III.
- class I region encodes HLA-A, B, C (‘classical’) antigens
- class II region encodes HLA-DR, DQ, DP antigens
- class III region encodes HSP70, TNF, C4A, C4B, C2, BF, CYP21
• Class I genes ~3-6kb
• Class II genes ~4-11kb (larger)
• Class I & II important in transplants

149
Q

What are class 1 MHC?

A

• Class I antigens: bind to Cytosolic pathogens (HLA-A, B, Cw) found on all nucleated cells- they present endogenous peptides form inside of cells. HLA CI A,B,C molecules composed of MHC encoded 45kd heavy chain non-covalently associated with non-polymorphic B2 microglobulin.

CD8

150
Q

What are class 2 MHC?

A

• Class II antigens: bind intravesicular pathogens and extracellular pathogens and toxins (HLA-DR, DQ, DP) primarily expressed on B lymphocytes but expression can be induced on T lymphocytes and other cells- exterior env (take things inside cell to present) HLA CII DR, DQ, DP molecules composed of MHC encoded 31-34 kd associated a chain non-covalently associated with 26-29 kd B chain

CD4

151
Q

What is the pattern of MHC inheritance?

A
Mendelian inheritance (1:4 chance of being mismatched, 1:2 sharing, 1;4 complete match)
•	 Inherit MHC en-bloc from each parental chromosome (HLA-A,B,Cw, DR, DQ,DP). Each individual inherits two antigens at a given locus.
•	 Codominant expression. All of the inherited antigens are displayed on the cell surface (inherited block called HLA phenotype).
152
Q

What is HLA typing?

A
  • Serological – cell based
  • Molecular
  • Extraction of DNA
  • Amplification
  • Detection of sequence polymorphisms (i.e. tissue type)
  • Hybridisation to probes
  • Sequencing
153
Q

Why do HLA typing?

A
  • Less rejection episodes
  • Better graft survival
  • National kidney waiting lists
  • Less sensitisation
  • Establish relationships
154
Q

Why detect antibodies before transplant?

A
  • Prevents hyperacute rejection
  • General – against many HLA types
  • Specific – against donor
  • Pretransplant crossmatch
  • Living or cadaveric
  • Avoids aborted transplant
  • Sensitising events
  • Previous transplant
  • Pregnancy
  • Blood transfusion
155
Q

What is the Complement Dependent Cytotoxicity Test ?

A
  • Developed in 1970s
  • Detects complement fixing IgG /IgM HLA and non-HLAs
  • Advantages
  • > 30yrs experience
  • Inexpensive
  • Disadvantages
  • Limited sensitivity
  • Subjective
  • Non-complement fixing abs
  • Viable reagent supply and quality control
  • Non HLA antibody interference
156
Q

What is the Interaction Between Donor & Recipient ? Indirect vs direct pathway?

A

• Indirect recognition pathway;
o Standard immunity
o APC take up antigen, peptides presented to immune system= attack what’s causing insult

• Direct recognition pathway;
o ONLY happens in transplant
o Donor APC cells migrate out of graft & interact with recipients white cells

157
Q

What are Causes of Sensitisation?

A
  • Immune response when exposed to foregin HLA, this could be through e.g. graft (e.g. a previous transplant), blood transfusions & female becomes sensitized during pregnancy (mum exposed to antigens in cells of baby)
  • Sensitization calc- if have pre-formed antibodies so sensitive to antigen
  • Cross match test; donor lymphocyte with HLA, interact donor cell & recip sreum (if it contains antibodies they will bind)- see if can get transplant
158
Q

What is Hypracute Rejection to Transplant?

A
  • Activates clotting cascade
  • Activate complement
  • Preformed antibodies
  • Xenograft rejection: Anti-Galα1-3Gal
  • ABO incompatible transplantation: ABO antibodies
  • HLA incompatible transplantation: Anti-HLA
159
Q

What is Acute Cellular Rejection?

A
cell dependent
•	Animal models
•	T cell immunosuppression
•	Directed against foreign HLA molecules
•	Effect of HLA mismatch
•	Typically 7- 10 days after transplant