Immunology Flashcards Preview

Laz's Pathology > Immunology > Flashcards

Flashcards in Immunology Deck (270)
Loading flashcards...
1
Q

What do sebaceous glands produce that has antibacterial effects?

A

Hydrophobic oils – repels water and microorganisms
Lysozyme – destroys the structural integrity of the bacterial cell wall
Ammonia and defensins – anti-bacterial properties

2
Q

How do NK cells determine whether to lyse cells or not?

A

They have inhibitory receptors which recognise self HLA and they have activating receptors that recognise heparan sulphate proteoglycans
The balance of these signals determines the response
They kill ‘altered self’ cells (e.g. malignancy or virus-infected cells)

3
Q

What does a dendritic cell do after phagocytosis?

A

Upregulate expression of HLA molecules
Express co-stimulatory molecules
Migrate via lymphatics to lymph nodes

4
Q

Which receptor is involved in the migration of dendritic cells to lymph nodes?

A

CCR7

5
Q

For each of the following subsets of CD4+ T cell, list their polarising factors and effector factors.

a. Th1
b. Th2
c. Th17
d. Follicular T cell
e. Treg

A
a.	Th1
Polarising
	IL-12
	IFN-gamma
Effector
	IL-2
	IL-10
	IFN-gamma
	TNF-alpha 
b.	Th2
Polarising
	IL-4
	IL-6
Effector
	IL-4
	IL-5
	IL-10
	IL-13
c.	Th17
Polarising
	IL-6
	TGF-beta
Effector
	IL-17
	IL-21
	IL-22
d.	Follicular T cell
Polarising
	IL-6
	IL-1
	TNF-alpha
Effector 
	IL-2
	IL-10
	IL-21
e.	Treg
Polarising
	TGF-beta
Effector
	IL-10
	Foxp3
	CD25
6
Q

Outline the mannose binding lectin pathway of complement activation.

A

Activated by the direct binding of MBL to microbial cell surface carbohydrates
This directly stimulates the classical pathway involving C4 and C2 (but NOT C1)
NOTE: this is NOT dependent on the adaptive immune response

7
Q

What are the effects of complement fragments that are released during complement activation?

A
Increase vascular permeability 
Opsonisation of immune complexes 
Opsonisation of pathogens 
Activation of phagocytes 
Promotes mast cell/basophil degranulation 
Punches holes in bacterial membranes
8
Q

What are the ligands for the CCR7 receptors on dendritic cells?

A

CCL19
CCL21
This interaction is important in directing dendritic cells towards lymph nodes

9
Q

Give three examples of failure of neutrophil production and outline their mechanism.

A

Reticular dysgenesis
 Autosomal recessive severe SCID with no production of lymphoid or myeloid cells
 Caused by failure of stem cells to differentiate along lymphoid or myeloid lineage
Kostmann syndrome
 Autosomal recessive congenital neutropaenia (mutation in HAX-1)
Cyclic neutropaenia
 Autosomal dominant episodic neutropaenia due to mutation in neutrophil elastase (ELA-2)
 Occurs every 4-6 weeks

10
Q

Describe the pathophysiology of leucocyte adhesion deficiency.

A

Caused by deficiency of CD18
CD18 normally combined with CD11a to produce LFA-1
LFA-1 normally binds to ICAM-1 on endothelial cells to mediate neutrophil adhesions and transmigration
A lack of CD18 means a lack of LFA-1, so neutrophils cannot enter tissues
During an infection, neutrophils will be mobilised from the bone marrow (HIGH neutrophils in the blood) but they will not be able to cross into the site of infection (NO pus formation)

11
Q

Outline the pathophysiology of chronic granulomatous disease.

A

Absent respiratory burst (deficiency of components of NADPH oxidase leads to inability to generate oxygen free radicals)
Excessive inflammation (persistent neutrophils and macrophage accumulation with failure to degrade antigens)
Granuloma formation
Lymphadenopathy and hepatosplenomegaly
Can be treated with IFN-gamma

12
Q

Describe the cytokine cycle between macrophages and T cells.

A

Macrophages produce IL12 which stimulates T cells, which then produce IFN-gamma
IFN-gamma acts back on the macrophages and stimulates the production of TNF-alpha and free radicals
Deficiencies in IL12, IL12R, IFN-gamma or IFN-gamma receptor can cause immunodeficiency (inability to form granulomas - mycobacterial)

13
Q

Name and describe the colour changes of two tests used to investigate chronic granulomatous disease.

A

Nitroblue Tetrazolium (NBT) – yellow to blue
Dihydrorhodamine (DHR) – fluorescent
NOTE: both of these tests are looking at the ability of neutrophils to produce hydrogen peroxide and oxidative stress

14
Q

What is the main clinical consequence of complement deficiency?

A

Increased susceptibility to infection by encapsulated bacteria

NOTE: Properidin (P) deficiency will also lead to increased risk of meningococcal infection

15
Q

Outline the clinical phenotype of complement deficiency.

A

SLE (if early components involved (e.g. C2)
Usually have severe skin disease
Increased risk of infection (common pathway deficiency)

16
Q

What are nephritic factors?

A

Autoantibodies that are directed against components of the complement pathway
They stabilise C3 convertases (break down C3) resulting in C3 activation and consumption

17
Q

What disease is associated with the presence of nephritic factors?

A

Membranoproliferative glomerulonephritis
It may also be associated with partial lipodystrophy

NOTE: it can cause a ‘tram track’ appearance on microscopy (immune complex and complement proteins deposit in the subendothelium

18
Q

Outline the management of complement deficiencies.

A

Vaccination (especially against encapsulated organisms)
Prophylactic antibiotics
Treat infection aggressively
Screen family members

19
Q

What is basophilic stippling?

A

Basophilic appearance of red blood cells caused by the presence of aggregated ribosomal material

20
Q

In which conditions might you see target cells (codocytes)?

A
Iron deficiency 
Thalassemia 
Hyposplenism 
Liver disease 
NOTE: target cells have a high SA: V ratio
21
Q

What are Howell-Jolly bodies? Which condition are they associated with?

A

Nuclear remnants present within red blood cells

Present in hyposplenism

22
Q

Which deficiencies are typically seen in Coeliac disease?

A
Iron 
B12 
Folate 
Fat 
Calcium
23
Q

Which HLA alleles are particularly common in patients with coeliac disease?

A

HLA-DQ2 (80%) – DQA10501 and DQB102 alleles

HLA-DQ8

24
Q

Describe the T cell response to gluten in coeliac disease.

A

Peptides from gluten (gliadin) are deamidated by tissue transglutaminase
Deamidated gliadin is taken up by antigen-presenting cells and presented via HLA molecules to CD4+ T cells
CD4+ T cell activation results in secretion of IFN-gamma and may increase IL-15 secretion
These cytokines promote activation of intra-epithelial lymphocytes (gamma-delta T cells)
The intraepithelial lymphocytes will kill epithelial cells via the NKG2D receptor (normally recognises the stress protein MICA)

NOTE: anti-gliadin antibodies are the most persistent

25
Q

List some other causes of high intraepithelial lymphocytes.

A
Dermatitis herpetiformis
Giardiasis
Cows’ milk protein sensitivity
IgA deficiency
Tropical sprue
Post-infective malabsorption
Drugs (NSAIDs)
Lymphoma
26
Q

How often should a DEXA scan be performed in coeliac patients?

A

Every 3-5 years

27
Q

Name a defect in stem cells that causes SCID and name the gene that is mutated.

A

Reticular dysgenesis – adenylate kinase 2 (AK2)

NOTE: this is a mitochondrial energy metabolism enzyme

28
Q

Which mutation is responsible for X-linked SCID?

A

Mutation in common gamma chain on Xq13.1
This is a component of many cytokine receptors (in particular, IL2) leading to an inability to respond to cytokines, causing arrest in T and NK cell development and the production of immature B cells

29
Q

Describe the typical cell counts you would expect to see in X-linked SCID.

A

Very low T cells
Very low NK cells
Normal or increased B cells
Low immunoglobulin

30
Q

Describe the typical cell counts you would expect to see in ADA deficiency.

A

Very low T cells
Very low B cells
Very low NK cells

31
Q

In which group of syndromes does the thymus gland fail to develop properly?

A

22q11.2 deletion syndromes (e.g. Di George syndrome)

This is characterised by failure of development of the pharyngeal pouch

32
Q

What are the immunological consequences of an underdeveloped thymus gland?

A

Normal B cell count
Low T cell count
Homeostatic proliferation with age (T cell numbers increase with age)
Immune function is mildly impaired and tends to improve with age

33
Q

Which defect leads to bare lymphocyte syndrome type 2?

A

Defects in the regulatory proteins involved in expression of class II genes:
• Regulatory factor X
• Class II transactivator

34
Q

List some investigations that may be used for suspected T cell deficiencies.

A

Total white cell count and differentials
Lymphocyte subsets
Immunoglobulins
Functional tests of T cell activation and proliferation
HIV test

35
Q

Describe the typical levels of CD4, CD8, B cells, IgM and IgG that you would expect to see in the following diseases:

a. SCID
b. Di George
c. BLS Type 2

A
a.	SCID 
CD4 low 
CD8 low 
B cells normal/low 
IgM normal/low 
IgG low 
b.	Di George
CD4 low 
CD8 low 
B cells normal 
IgM normal 
IgG normal/low 
c.	BLS Type 2
CD4 low 
CD8 normal 
B cells normal 
IgM normal 
IgG low
36
Q

Outline the pathophysiology of Bruton’s X-linked hypogammaglobulinaemia.

A

Prevents the maturation of B cells at that point at which they emerge from the bone marrow
Caused by an abnormal B cell tyrosine kinase (BTK) gene
This results in the absence of mature B cells and, hence, an absence of antibodies

37
Q

Outline the pathophysiology of X-linked hyper IgM syndrome.

A

Blocks the maturation of IgM B cells through germinal centres into B cells that produce other classes of immunoglobulin (i.e. prevents germinal centre reactions)
Caused by a mutation in the CD40 ligand gene
This is technically a T cell problem, however, it means that CD4+ T helper cells cannot provide help to B cells so they cannot undergo germinal centre reactions
NOTE: CD40 ligand is encoded on Xq26

38
Q

What is common variable immunodeficiency and what are the main features?

A

A group of disorders caused by some form of failure of differentiation of B lymphocytes
Defined by:
• Marked reduction in IgG, IgA and IgE
• Poor/absent response to immunisation
• Absence of other defined immunodeficiency

39
Q

List some investigations that may be used for suspected B cell deficiencies.

A

Total white cell count and differential
Lymphocyte subsets
Serum immunoglobulins and protein electrophoresis
Functional tests of B cell function (e.g. measure IgG antibody against a specific pathogen (e.g. S. pneumoniae), if this is low, vaccinate using a killed vaccine and check levels again in 6-8 weeks)
NOTE: IgG production is a surrogate marker for CD4+ T helper cell function

40
Q

Which protein is upregulated in autoinflammatory diseases caused by a gain-of-function mutation in NLRP3? Name 3 diseases that are caused by this mutation.

A

Cryopyrin (NALP3)
Muckle Wells syndrome
Familial cold autoinflammatory syndrome
Chronic infantile neurological cutaneous articular syndrome
All of these are autosomal dominant
NOTE other examples of monogenic autoinflammatory conditions: TNF receptor associated periodic syndrome (TNF receptor mutation), Hyper IgD with periodic fever syndrome (mevalonate kinase mutation)

41
Q

Which gene mutation causes Familial Mediterranean Fever and which protein does this gene encode?

A

MEFV gene

Encodes pyrin-marenostrin which is a negative regulator of the inflammatory pathway

42
Q

Describe how the inflammasome complex functions.

A

The pathway is activated by toxins, pathogens and urate crystals
These act via cryopyrin and ASC (apoptosis-associated speck-like protein) to activate procaspin 1
Activation of procaspin 1 results in the production of NFB, IL1 and apoptosis
Pyrin-maronestrin is a negative regulator of this pathway

NOTE: pyrin-marenostrin is found in neutrophils

43
Q

What is the inheritance pattern of Familial Mediterranean Fever?

A

Autosomal recessive

44
Q

Outline the clinical presentation of Familial Mediterranean Fever.

A
Periodic fevers lasting 48-96 hours associated with
•	Abdominal pain (peritonitis)
•	Chest pain (pleurisy, pericarditis)
•	Arthritis 
•	Rash

It can lead to AA amyloidosis

45
Q

Outline the treatment of Familial Mediterranean Fever.

A

Colchicine 500 µg BD (binds to tubulin and disrupt neutrophil migration and chemokine secretion)
2nd line: blocking cytokines
• Anakinra – IL1 receptor blocker
• Etanercept – TNF-alpha blocker

46
Q

Which autoimmune conditions tend to occur in APECED?

A
Hypoparathyroidism (COMMON) 
Addison’s disease (COMMON)
Hypothyroidism 
Diabetes mellitus 
Vitiligo 

APECED: autoimmune polyendocrinopathy candidasis ectodermal dystrophy (aka polyglandular autoimmune syndrome type 1)

47
Q

Why are patients with APECED prone to Candida infections?

A

They produce antibodies against IL17 and IL22

48
Q

What does IPEX stand for?

A

Immune dysregulation polyendocrinopathy enteropathy X-linked syndrome

Caused by mutation in FoxP3 (Forkhead Box P3)

49
Q

Which mutations cause ALPS?

A

Mutations in the FAS pathway leading to defects in apoptosis of lymphocytes
This leads to a failure of lymphocyte tolerance (as autoreactive lymphocytes don’t die by apoptosis) and failure of lymphocyte homeostasis (you keep producing lymphocytes)

50
Q

Describe the clinical phenotype of ALPS.

A

High lymphocyte count
Large spleen and lymph nodes
Autoimmune disease (usually cytopaenias)
Lymphoma

51
Q

What is the best known chromosomal region that is implicated in Crohn’s disease?

A

IBD1 on chromosome 16 (NOD2/CARD15 gene)

NOTE: NOD2 is found in the cytoplasm of myeloid cells and is a microbial sensor, mutation is also seen in Blau syndrome

NOD2 = nucleotide-binding oligomerization domain-containing protein 
CARD15 = caspase-activating recruitment domain
52
Q

List the autoimmune diseases associated with the following HLA polymorphisms:

a. DR3
b. DR3/4
c. DR4
d. DR15

A
a.	DR3
Graves’ disease 
SLE 
b.	DR3/4
Type 1 diabetes mellitus 
c.	DR4
Rheumatoid arthritis 
d.	DR15
Goodpasture’s syndrome
53
Q

Name and state the function of 2 genes that are involved in T cell activation and are often mutated in polygenic autoimmune disease.

A

PTPN22 – suppresses T cell activation
CTLA4 – regulates T cell function (expressed by T cells)

PTPN = protein tyrosine phosphate non-receptor 22
CTLA = cytotoxic T lymphocyte-associated protein 4
54
Q

Name the autoantigen in the following diseases:

a. Goodpasture’s disease
b. Pemphigus vulgaris

A

a. Goodpasture’s disease
Alpha-3 subunit of non-collagenous domain of collagen IV
b. Pemphigus vulgaris
Epidermal cadherin

55
Q

Describe the difference between immune responses mediated by Th1 and Th2 cells.

NOTE: both are types of CD4 cell

A
Pathogens that have conserved structures (PAMPs) such as bacteria are recognised by Th1 and Th17 cells 
Multicellular organisms (e.g. helminths) and allergens don’t have conserved structured but they release mediators that damage epithelial cells. Disturbance of epithelial cells is recognised by the Th2 cells
56
Q

List some elective investigations for allergic disease.

A
Skin prick and intradermal tests 
Specific IgE measurement 
Component resolved diagnostics 
Basophil activation test 
Challenge test
57
Q

What is component resolved diagnostics?

A

A blood test to detect IgE to single protein components (useful for peanut and hazelnut allergy)
IgE sensitisation to heat and proteolytic labile proteins = minor symptoms
IgE sensitisation to heat and protolytic stable protein = major symptoms

58
Q

List some indications for allergy component testing.

A

Detect primary sensitisation
Confirm cross-reactivity
Define risk of serious reaction for stable allergens

59
Q

What is a basophil activation test?

A

Measurement of basophil response to allergen IgE cross-linking
Activated basophils show increased expression of CD63, CD203 and CD300
This is increasingly used in food and drug allergy

60
Q

List some mechanisms of anaphylaxis.

A

IgE – mast cells and basophils – histamine and PAF (triggered by food, venom, ticks, penicillin)
IgG – macrophages and neutrophils – histamine and PAF (triggered by blood product transfusions)
Complement – mast cells and macrophages – histamine and PAF (triggered by lipid excipients, liposomes, dialysis membranes)
Pharmacological – mast cells – histamine and leukotrienes (triggered by NSAIDs)

61
Q

List some IgE-mediated food allergy syndromes.

A

Anaphylaxis (e.g. peanut)
Food-associated exercise-induced anaphylaxis (ingestion of food leads to anaphylaxis if the individual exercises within 4-6 hours of ingestion (e.g. wheat, shellfish))
Delayed food-induced anaphylaxis to beef/pork/lamb (symptoms occur 3-6 hours after ingestion, induced by tick bites)
Oral allergy syndrome (limited to oral cavity with swelling and itching, occurs after pollen allergy is established, caused by cross-reaction of IgE antibody to pollen with stone fruits (e.g. apples), vegetables and nuts)

62
Q

Which chromosome is HLA encoded on?

A

Chromosome 6

63
Q

Describe the basic structure of HLA Class I and Class II.

A

Class I: have three alpha domains and a beta-2 microglobulin domain, has one transmembrane domain
Class II: has two alpha and two beta domains, had two transmembrane domains

64
Q

Which HLA alleles are most immunogenic?

A

A, B and DR

65
Q

What are the typical histological features of T-cell mediated rejection?

A

Lymphocytic interstitial infiltration
Ruptured tubular basement membrane
Tubulitis (inflammatory cells within the tubular epithelium)

66
Q

Name and describe three assays for anti-HLA antibodies before transplants.

A
Cytotoxic Assays (CDC): tests whether patient serum kills donor lymphocytes in the presence of complement 
Flow Cytometry (FACS): tests whether patient serum binds donor lymphocytes irrespective of complement 
Solid Phase Assays (Luminex): beads containing all the possible HLA epitopes are mixed with the patient’s serum. This determines which HLA types the patient has antibodies against. Having many antibodies against different HLA epitopes suggests that the patient is highly sensitised.
67
Q

Name two drugs that target TCR.

A

Anti-CD3 antibody (OKT3 - also known as muromonab-CD3)
Anti-thymocyte globulin

Used for transplant rejection

68
Q

Name an anti-CD52 antibody and state its effect.

A

Alemtuzumab (campath) – causes lysis of T cells

Used for solid organ transplantation, multiple sclerosis and NHL, CLL, MS
Side-effects: ITP, Graves disease

69
Q

Name an anti-CD25 antibody and state its effect.

A

Daclizumab – targets cytokine signalling

70
Q

How to BAFF inhibitors work?

A

Target cytokines (BAFF) that promote B cell activation and growth

71
Q

Name a proteasome inhibitor and describe how it works.

A

Bortezomib

Blocks the production of antibodies by plasma cells

72
Q

Name a complement inhibitor.

A

Eculizumab

73
Q

Outline the components of modern transplant immunosuppression regimes.

A

Induction agent (e.g. OKT3, anti-CD52, anti-CD25)
Baseline immunosuppression (e.g. calcineurin inhibitor, mycofenolate mofetil, azathioprine, steroids)
Treatment of acute rejection
• Cellular: steroids, OKT3
• Antibody-Mediated: IVIG, plasma exchange, anti-CD20

74
Q

Which antibodies have a protective role in HIV infection?

A

Anti-gp120
Anti-gp41 (Nt)
Non-neutralising anti-p24 gag IgG (CD4+ cells recognise these antigens when presented on MHC class II)
NOTE: HIV remains infectious even when coated with antibodies

75
Q

What are the screening and confirmatory tests for HIV?

A

Screening: HIV antibody ELISA
Confirmatory: HIV antibody Western blot

76
Q

Describe how haemagglutinin inhibition assays work.

A

If you put normal red blood cells in a petri dish, they will clump at the bottom forming a red spot
If you add influenza virus, the HA makes red cells stick together and causes a diffuse coloration across the well
If you add the serum of someone who has a lot of antibodies against HA, it will inhibit the haemagglutination effects of HA so the red cells remain as a discrete red spot
The higher the dilution of serum at which the red cells remain as a little dot, the more antibodies are present in the serum
NOTE: sialic acid receptors on RBCs bind to HA leading to haemagglutination

77
Q

What is a live attenuated virus vaccine? List some examples.

A

The organism is alive but modified to limit its pathogenesis
Examples: MMR, typhoid, BCG, yellow fever, polio (Sabin)

78
Q

List some examples of the following types of vaccine:

a. Toxoids
b. Component/Subunit

A
a.	Toxoids
Diphtheria
Tetanus
b.	Component/Subunit 
Hep B (HBsAg)
HPV (capsid)
Influenza (HA)
79
Q

Describe how conjugate vaccines work.

A

Polysaccharide and protein carrier
Polysaccharide induces a T-cell independent B cell response (transient)
Addition of the protein carrier promoted T cell immunity which enhances B cell/antibody responses

80
Q

List some examples of conjugate vaccines.

A

Haemophilus influenzae type B
Meningococcus
Pneumococcus

81
Q

List some indications for IVIG.

A
Primary antibody defect 
•	X-linked agammaglobulinaemia
•	X-linked hyper IgM syndrome 
•	Common variable immunodeficiency
Secondary antibody defect
•	CLL
•	Multiple myeloma 
•	After bone marrow transplantation
82
Q

List four types of T cell adoptive cell transfer.

A

Virus-specific T cells
Tumour infiltrating T cells (TIL)
T cell receptor T cells (TCR)
Chimeric antigen receptor T cells (CAR T Cell Therapy)

83
Q

Using an example, describe how virus-specific T cells are used.

A

Used for EBV in patients who are immunosuppressed to prevent the development of lymphoproliferative disease
Blood is taken from the patient or from a donor
Peripheral blood mononuclear cells are isolated and stimulated with EBV peptides
This creates an expansion of EBV-specific T cells which are then reinfused into the patient
NOTE: tumour infiltration T cell therapy follows the same principle but uses tumour antigens

84
Q

Describe how TCR and CAR T cell therapy works.

A

T cells are taken from the patient and vectors are used to insert gene fragments that encode receptors
In TCR therapy, the gene will encode a specific TCR (e.g. against tumour antigen)
In CAR therapy, the receptors are chimeric (containing both B and T cell components)

85
Q

Describe a use of CAR T cell therapy.

A

Used to target CD19 (present on B cells)
Receptors on the CAR cell have an immunoglobulin variable domain and is joined to a TCR
This means that it recognises CD19 through an immunoglobulin domain but signals through the TCR pathway
NOTE: this is used in ALL and NHL

86
Q

What is ipilimumab and how does it work?

A

CTLA4 and CD28 are both expressed by T cells and they recognise antigens (CD80 and CD86) on APCs
Signalling through CD28 results in a stimulatory response
Signalling through CTLA4 results in an inhibitory response
Ipilimumab is a monoclonal antibody that blocks CTLA4 thereby removing this inhibitory response
It is used in advanced melanoma

87
Q

Explain the use of antibodies against PD-1 in treating cancer.

A

PD-1 and PD-2 ligands are present on APCs and interact via PD-1 receptors on T cells to cause an inhibitory response
They can also be expressed by some tumour cells
Pembrolizumab and nivolumab are antibodies that are specific to PD-1, thereby blocking this effect
This is also used in advanced melanoma

88
Q

List some examples of the therapeutic use of recombinant cytokines.

A

Interferon alpha – used as an adjunct in the treatment of Hep B, Hep C, Kaposi sarcoma, CML and multiple myeloma
Interferon beta – Behcet’s disease, relapsing MS
Interferon gamma – chronic granulomatous disease

89
Q

Describe the effects of steroids on:

a. Prostaglandins
b. Phagocytes
c. Lymphocyte Function

A

a. Prostaglandins
Inhibits phospholipase A2
Phospholipase A2 is responsible for the conversion of phospholipids into arachidonic acid (which will then be converted to eicosanoids by COX)
Inhibiting phospholipase A2 leads to a reduction in arachidonic acid and prostaglandin formation and, hence, a reduction in inflammation
b. Phagocytes
Decrease traffic of phagocytes to inflamed tissue (reduces the expression of adhesion molecules on the endothelium)
This leads to a transient increase in neutrophil count
Decreased phagocytosis
Decreases proteolytic enzymes
c. Lymphocyte Function
Lymphopaenia (sequestration in lymphoid tissue)
Blocks cytokine gene expression
Decreased antibody production
Promotes apoptosis

90
Q

List some examples of anti-proliferative agents.

A

Cyclophosphamide
Mycophenolate
Azathioprine
Methotrexate

91
Q

What is the mechanism of action of cyclophosphamide?

A

Alkylates the guanine base of DNA which damages the DNA and prevents replication
Affects B cells more than T cells

92
Q

List some side-effects of cyclophosphamide.

A

Toxic to proliferating cells – bone marrow suppression, sterility (mainly males), hair loss
Haemorrhagic cystitis – due to toxic metabolic (acrolein) in the urine
Malignancy – bladder cancer, haematological malignancy, non-melanoma skin cancer
Teratogenic
Infection (e.g. PCP)

93
Q

Outline the mechanism of action of azathioprine.

A

Metabolised by the liver to 6-mercaptopurine
Blocks de novo purine synthesis (e.g. adenine and guanine)
Prevents DNA replication
Preferentially inhibits T cell activation and proliferation

94
Q

Which precaution must you take before starting a patient on azathioprine?

A

Check TPMT activity – 1 in 300 individuals have a TPMT polymorphism which means that they are unable to metabolise azathioprine leading to bone marrow suppression

95
Q

Outline the mechanism of action of mycophenolate mofetil.

A

Blocks de novo nucleotide synthesis
Prevents replication of DNA
Affects T cell proliferation more than B cells
Non-competitive inhibitor of IMPDH (inosine-5-monophosphate dehydrogenase)

96
Q

List some side-effects of mycophenolate mofetil.

A

Bone marrow suppression
Teratogenic
Infection (particularly HSV reactivation and PML (JC virus))

97
Q

Describe the mechanism of action of calcineurin inhibitors.

A

Normally, TCR engagement leads to increased cytoplasmic calcium which binds to calmodulin leading to the activation of calcineurin
Calcineurin then activates NFATc resulting in the upregulation of IL2
Calcineurin inhibitors block this pathway, thereby blocking IL2 production

98
Q

What are the main side-effects of calcineurin inhibitors?

A

Hypertension and nephrotoxicity
Diabetes
Neurotoxic
Gingival hypertrophy (ciclosporin)

99
Q

Give an example of a JAK inhibitor. State which disease it is used to treat.

A

Tofacitinib (JAK1 and JAK2 inhibitor)

Rheumatoid arthritis

100
Q

Give an example of a PDE4 inhibitor. State which disease it is used to treat.

A

Apremilast

Psoriasis

101
Q

Describe the mechanism of action of PDE4 inhibitors.

A

PDE4 is important in the metabolism of cAMP
PDE4 inhibitors result in increased levels of cAMP which activates PKA and prevents the activation of transcription factors
This leads to a decrease in cytokine production
Effective in psoriasis and psoriatic arthritis

102
Q

For each of the following monoclonal antibodies, state the antigen that they are targeting:

a. Basiliximab
b. Abatacept
c. Rituximab
d. Natalizumab
e. Tocilizumab

A
a.	Basiliximab
Anti-CD25
b.	Abatacept
CTLA4-Ig 
c.	Rituximab
Anti-CD20
d.	Natalizumab
Anti-4 integrin 
e.	Tocilizumab
Anti-IL6 receptor
103
Q

List some side-effects of anti-thymocyte globulin.

A

Infusion reactions
Leukopaenia
Infection
Malignancy

104
Q

Describe the mechanism of action and the use of basiliximab.

A

Targets IL2 receptor alpha chain (aka CD25)
This part of the receptor is specific for IL2 receptors
Results in inhibition of T cell proliferation

Uses: allograft rejection

105
Q

Describe the mechanism of action of abatacept.

A

It is made from the fusion of CTLA4 and IgG Fc
APCs bind to CTLA4 (inhibitory) and CD28 via CD80 and CD86 receptors
Abatacept binds to CD80 and CD86 receptors and prevents engagement with T cells thereby reducing T cell activation
It is effective in rheumatoid arthritis

106
Q

List some indications for rituximab.

A

Lymphoma
Rheumatoid arthritis
SLE
NOTE: it is given as two IV doses every 6-12 months

107
Q

Describe the mechanism of action of natalizumab.

A

Antibody against alpha-4 integrin
Alpha-4 integrin is expressed with beta-1 or beta-7
This complex binds to VCAM1 or MadCAM1 to mediate rolling and arrest of leukocytes
Blocking this integrin inhibits leukocyte migration
Uses: multiple sclerosis, Crohn’s disease

108
Q

Describe the mechanism of action of tocilizumab.

A

Antibody against IL6 receptor

Results in reduced activation of macrophages, T cells, B cells and neutrophils

109
Q

What are the main indications of tocilizumab?

A

Castleman’s disease (IL6-producing tumour)

Rheumatoid arthritis

110
Q

Which antigens are targeted by the following monoclonal antibodies:

a. Ustekinumab
b. Secukinumab
c. Denosumab

A
a.	Ustekinumab
IL12
IL23
b.	Secukinumab
IL17 
c.	Denosumab
RANKL 

NOTE: menopause stimulates RANKL production

111
Q

Describe the mechanism of action of etanercept.

A

It is a decoy receptor that mops up TNF alpha thereby inhibiting its action
It is given as an SC injection
Used in rheumatoid arthritis, psoriasis and ankylosing spondylitis

112
Q

Describe the mechanism of action of ustekinumab.

A

Antibody against the p40 subunit that is found in IL12 and IL23
This affects Th17 activation and IL17 signalling
These cytokines mainly act on NK cells and T cells thereby modulating their activity
Used in psoriasis and Crohn’s disease

113
Q

Describe the mechanism of action and uses of secukinumab.

A

Antibody to IL17A thereby inhibiting its effect

Indications include psoriasis and ankylosing spondylitis

114
Q

Describe the mechanism of action of denosumab.

A

RANKL is produced by osteoblasts and it acts on RANK receptors on osteoclasts
It promotes osteoclast differentiation and function, thereby leading to increased bone resorption
Osteoprotegrin is a natural decoy receptor for RANKL which regulates the system
Denosumab binds to RANKL and reduces osteoclast differentiation and function
NOTE: it is used for osteoporosis and is administered as SC injections every 6 months

115
Q

List some autoantibodies that are found in type I diabetes mellitus.

A
Anti-GAD 
Anti-IA2
Anti-islet cell 
Anti-insulin 
NOTE: the detection of these antibodies does not currently play a part in diagnosis of diabetes mellitus
116
Q

List some genetic polymorphisms that predispose to rheumatoid arthritis.

A
HLA DR1
HLA DR4
PTPN22
PAD 2 and PAD 4 polymorphisms
Polymorphisms affecting TNF, IL1, IL6 and IL10
117
Q

What is a key common feature amongst HLA alleles that are associated with rheumatoid arthritis?

A

They share a sequence at position 70-74 of the HLA DR-beta chain (shared epitope)
This enables binding of HLA to arthritogenic peptides (particularly citrullinated peptides)

118
Q

Describe the role of PAD in the pathogenesis of rheumatoid arthritis.

A
Peptidylarginine deaminases (2 and 4) are involved in the deamination of arginine to form citrulline 
Polymorphisms that are associated with increased citrullination leads to a high load of citrullinated peptides
119
Q

List some environmental factors that contribute to the pathogenesis of rheumatoid arthritis.

A

Smoking is associated with the development of erosive disease (due to increased citrullination)
Gum infection by Porphyromonas gingivalis is associated with rheumatoid arthritis as it expresses PAD, thereby promoting citrullination

120
Q

Describe B cell involvement in the pathophysiology of rheumatoid arthritis.

A

Type II – antibodies bind to citrullinated peptides leading to activation of macrophages, NK cells and complement
Type III – immune complexes form and get deposited leading to complement activation

121
Q

Outline the roles of T cells in the pathophysiology of rheumatoid arthritis.

A

APCs present peptides to CD4+ T cells which then produce IFN-gamma and IL17
These cytokines act on fibroblasts and macrophages
This leads to the production of MMPs, IL1 and TNF-alpha

122
Q

Which type of hypersensitivity reaction is SLE?

A

Type III hypersensitivity – antibodies bind to antigens forming immune complexes which deposit in tissues (e.g. skin, joints, kidneys) and activated complement via the classical pathway
These antibodies can also stimulate cells that express Fc receptors

123
Q

What are the two types of ANA and what are their immunofluorescence patterns?

A

Anti-dsDNA – homogenous staining pattern, they are highly specific for SLE and high titres are associated with severe disease (useful for disease monitoring)
Anti-ENA4 (extractable nuclear antigens such as ribonucleoproteins (e.g. Ro, La, Sm)) - speckled immunofluorescence

124
Q

Which antibodies are tested for in antiphospholipid syndrome?

A

Anti-cardiolipin antibody – immunoglobulins directed against phospholipids and 2 glycoprotein-1
Lupus anticoagulant – prolongation of phospholipid-dependent coagulation tests.
NOTE: cannot be assessed if the patient is on anticoagulant therapy
NOTE: both tests should be performed as 40% of patients have disconcordant antibodies

125
Q

Which cells are particularly important in the pathophysiology of systemic sclerosis?

A

Th2 and Th17

NOTE: TGF-beta, produced by macrophages and T cells, is important in stimulating the deposition of collagen

126
Q

Describe the differences between the histology of dermatomyositis and polymyositis.

A

Dermatomyositis – perivascular CD4+ T cell and B cells are seen, this can cause an immune complex-mediated vasculitis (type III response)
Polymyositis – CD8+ T cells surround HLA Class I expressing myofibres, CD8+ T cells kill these myofibres via granzyme/perforin (type IV response)

127
Q

Which antibodies are seen in dermatomyositis and polymyositis?

A

Dermatomyositis: anti-aminoacyl tRNA synthetase (e.g. Jo-1), anti-Mi2
Polymyositis: anti-signal recognition peptide (anti-SRP) antibody

NOTE: anti-aminoacyl tRNA synthetase antibody is cytoplasmic

128
Q

Outline the pathophysiology of ANCA.

A

These antibodies are specific to antigens located within primary granules within the cytoplasm of neutrophils
Inflammation may lead to expression of these antigens on the surface of neutrophils
Antibody engagement with these antigens may lead to neutrophil activation (type II hypersensitivity)
NOTE: these are different from anti-nuclear antibodies

129
Q

Describe the key difference between cANCA and pANCA.

A

cANCA
• Cytoplasmic fluorescence
• Associated with antibodies against proteinase 3
• Occurs in > 90% of Wegener’s patients with renal involvement
pANCA
• Perinuclear staining pattern
• Associated with antibodies to myeloperoxidase
• Less sensitive and specific than cANCA
• Associated with MPA and Churg-Strauss syndrome

130
Q

Describe the reactions involved in oxidative killing of pathogens within phagolysosomes.

A

NADPH oxidase converts oxygen into reactive oxygen species (e.g. superoxide and hydrogen peroxide)
Myeloperoxidase catalyses the production of hydrochlorous acid (from hydrogen peroxide and chloride)

131
Q

What are the polarising and effector factors of Th1?

A
Polarising
	IL-12
	IFN-gamma
Effector
	IL-2
	IL-10
	IFN-gamma
	TNF-alpha
132
Q

What are the polarising and effector factors for Th2?

A
Polarising
	IL-4
	IL-6
Effector
	IL-4
	IL-5
	IL-10
	IL-13
133
Q

What are the polarising and effector factors for Th17?

A
Polarising
	IL-6
	TGF-beta
Effector
	IL-17
	IL-21
	IL-22
134
Q

What are the polarising and effector factors for follicular T cells?

A
Polarising
	IL-6
	IL-1 beta
	TNF-alpha
Effector 
	IL-2
	IL-10
	IL-21
135
Q

What are the polarising and effector factors for Tregs?

A
Polarising
	TGF-beta
Effector
	IL-10
	Foxp3
	CD25
136
Q

Which types of infection tend to occur in patients with phagocyte deficiency?

A

Recurrent skin and mouth infections
 Bacteria – Staphylococcus aureus, enteric bacteria
 Fungi – Candida albicans, Aspergillus fumigatus
Mycobacterial infections (particularly with IL12 deficiency)
 TB, atypical mycobacteria

137
Q

Outline the treatment of NK cell deficiency.

A

Prophylactic antiviral drugs (e.g. aciclovir)
Cytokines (e.g. IFN-alpha to stimulate NK cytotoxic function)
Haematopoietic stem cell transplantation

138
Q

What is factor H?

A

A control protein in the complement cascade

139
Q

What causes hereditary angioedema?

A

C1 esterase inhibitor deficiency

It is autosomal DOMINANT

140
Q

List some complications of coeliac disease.

A

Malabsorption
Osteomalacia and osteoporosis
Neurological disease (epilepsy and cerebral calcification)
Lymphoma (causes multi-focal T cell lymphoma)
Hyposplenism

141
Q

What are the main clinical features of 22q11.2 deletion syndromes?

A

Facial abnormalities (high forehead, low set ears, cleft palate, small mouth and jaw)
Underdeveloped parathyroid gland (resulting in hypocalcaemia)
Oesophageal atresia
Underdeveloped thymus
Complex congenital heart disease

142
Q

What is the most common allergic disease in:
A) Children
B) Adults

A

A) food allergy (e.g. egg and milk)

B) allergic rhinitis

143
Q

List some triggers for the following types of mast cell degranulation:
A) IgE-mediated
B) non-IgE mediated

A

A) peanut, penicillin, wasp/bee venom, latex

B) NSAIDs, opioids, IV contrast, exercise

144
Q

Which autoimmune conditions are often seen in IPEX?

A

Enteropathy
Diabetes mellitus (Type 1)
Hypothyroidism
Dermatitis (eczema)

145
Q

What are three forms of peripheral tolerance?

A

T cell require costimulation to become activates (costimulatory molecules are upregulated in infection and inflammation)
Regulatory T cells
Immune privileged sites

146
Q

How should acute angioedema be treated?

A

IM adrenaline

147
Q

Which complement factor is important for chemotaxis?

A

C3a

148
Q

Which protein does the gene mutation that causes X-linked SCID encode?

A

IL-2 receptor

149
Q

What is Wiskott-Aldrich syndrome and which gene mutation causes it?

A

X-linked recessive condition characterised by eczema, thrombocytopaenia, immunodeficiency, bloody diarrhoea and nosebleeds
Low IgM, normal IgG, high IgA and IgE
Caused by WASP mutation

150
Q

What does the MHC Class III gene cluster encode?

A

C2
C4
B

151
Q

Which types of infection are people with IgA deficiency prone to?

A

Respiratory and gastrointestinal tract infections

NOTE: IgA deficiency is the only B cell maturation defect that responds to vaccines

152
Q

What is a major risk factor for chronic allograft rejection?

A

Hypertension

Hyperlipidaemia is also a risk factor

153
Q

How is acute rejection treated?

A

Antibody-mediated: IVIG and plasmapheresis
Cell-mediated: high dose corticosteroids
Hyperacute (preformed antibody): can only be prevented by accurate HLA-typing

154
Q

Which HLA types are most important in rejection?

A

HLA DR > B > A

NOTE: CD4 recognises DR; CD8 recognises A

155
Q

What are the main side-effects of methotrexate?

A

Pneumonitis
Pulmonary fibrosis
Cirrhosis

156
Q

What is the main side-effect of IVIG?

A

Anaphylaxis

157
Q

Describe the mechanism of action of alum as an adjuvant to vaccines.

A

Promotes a predominantly antibody response through the release of IL-4 that primes naïve B-cells

158
Q

Name a vaccine that is made up of recombinant protein?

I.e. using recombinant DNA technology

A

Hepatitis B vaccine

159
Q

Describe the typical presentation of SCID.

A
Unwell by 3 months 
Infections of all types 
Failure to thrive 
Persistent diarrhoea 
unusual skin disease
160
Q

List some features of Di George syndrome.

A
CATCH-22
Cardiac abnormalities 
Abnormal facies (high forehead, low set ears) 
Thymic aplasia
Cleft palate 
Hypocalcaemia/hypoparathyroidism 
22q11.2
161
Q

What are the clinical features of common variable immuondeficiency?

A

Recurrent bacterial infection (e.g. bronchiectasis, persistent sinusitis, recurrent GI infection)
Autoimmune disease
Granulomatous disease

162
Q

Which test result would be consistent with Factor B deficiency?

A

Low AP50

AP50 tests the activity of B, D, Properidin, C3 and C5-9

163
Q

Which foods are involved in latex food syndrome?

A
Chestnut 
Avocado 
Banana
Potato
Tomato
Kiwi
164
Q

What counts as a positive result on skin prick testing?

A

Wheal > 2 mm greater than the negative control

NOTE: discontinue antihistamines 48 hours before test, positive control is histamine, negative control is dilutent
Specific IgE is less sensitive but used if unable to stop antihistamines, history of anaphylaxis or extensive eczema

165
Q

What is the gold standard test for diagnosing food allergy?

A

Challenge test

But this carries a risk of severe reaction

166
Q

What investigation can be useful in patients without a convincing clinical diagnosis of anaphylaxis?

A

Mast cell tryptase (peaks at 1-2 hours and returns to baseline by 6 hours)

167
Q

What is Evan syndrome?

A

AIHA + ITP

168
Q

What antigen is targeted in ITP?

A

Glycoprotein IIb/IIIa

169
Q

Which antigens are antibodies developed against in rheumatic fever?

A

M proteins on Group A streptococcus

170
Q

What is mixed essential cryoglobulinaemia?

A

IgM against IgG (and sometimes hepatitis C antigens)
Causes joint pain, splenomegaly, skin, nerve and kidney involvement
Associated with hepatitis C

171
Q

Which antibodies are associated with Sjogren syndrome?

A

Anti-Ro
Anti-La
Speckled immunofluorescence
NOTE: classically diagnosed using Shirmer’s test

172
Q

Which antibodies are seen in autoimmune hepatitis?

A

Anti-smooth muscle antibodies (ASMA)
Anti-LKM1
Anti-SLA

173
Q

Which antibodies are seen in systemic sclerosis?

A

Limited: anti-centromere
Diffuse: topoisomerase/Scl70, RNA polymerase I/II/III, fibrillarin (nucleolar pattern)

174
Q

How often does human normal immunoglobulin need to be given as antibody replacement therapy?

A

Every 3-4 weeks

E.g. used in combined variable immunodeficiency, Bruton’s

175
Q

List some uses of interferon alpha, beta and gamma.

A

Alpha: hepatitis C, hepatitis B, Kaposi sarcoma, Hairy cell leukaemia, CML, myeloma
Beta: Behcet’s disease
Interferon gamma: chronic granulomatous disease

176
Q

What are the limitations of plasmapheresis?

A

Anaphylaxis

Rebound antibody production

177
Q

What is the mechanism of action of sirolimus?

A

Inhibits mTOR thereby blocking T cell replication

NOTE: mTOR inhibits IL2 signalling pathways

178
Q

What is the mechanism of action of Efalizumab?

A

Anti-CD11a

Inhibits migration of T cells

179
Q

What is the difference between direct and indirect recognition of a transplanted graft?

A

Direct - DONOR APC presents antigen to recipient T cells (responsible for acute rejection - everything is perceived as ‘foreign’)
Indirect - RECIPIENT APC presents donor antigen to recipient T cells (responsible for chronic rejection)

180
Q

Describe the symptoms of graft-versus-host disease.

A

Skin: rash
Gut: nausea, vomiting, abdominal pain, diarrhoea
Liver: jaundice

181
Q

Which cytokines do CD8 T cell produce that can prevent HIV entry into cells?

A

MIP-1a
MIP-1b
RANTES

These cytokines block co-receptors

182
Q

What is the median time from HIV infection to AIDS?

A

8-10 years

Rapid progressors: 2-3 years
Long term non-progressors: stable CD4 and asymptomatic after 10 years

183
Q

List some examples of NRTIs.

A

Zidovudine
Abacavir
Emtricitabine
Lamivudine

NOTE: tenofovir is a nucleoTide reverse transcriptase inhibitor

184
Q

List some examples of NNRTIs.

A

Nevirapine

Efavirenz

185
Q

List some examples of integrase inhibitors.

A

Raltegravir

Elvitegravir

186
Q

List some examples of protease inhibitors used for HIV.

A

Indinavir
Lopinavir
Saquinavir

187
Q

What are some key features of central memory cells?

A

Found in lymph nodes and extravasate in high endothelial venules
CCR7 and CD62L positive
Produce IL2
Central memory predominates in CD4 cells

188
Q

What are some key features of effector memory cells?

A

Found in liver, lungs and gut
CCR7 and CD62 NEGATIVE (not found in lymph nodes)
Produce perforin and IFN-gamma
Predominates in CD8 cells

189
Q

Why dont vaccines work very well in the elderly?

A

Immune senescence

190
Q

List some adjuvants that can be used with vaccinations.

A

Alum: most common. Allows slow release of antigens. Activates Gr1 cells to produce IL4 (primes B cells)
CpG: linked to DNA motif rich in CpG. Activates TLRs on APCs by stimulating costimulatory molecule expression
Freund’s adjuvant: water-in-oil emulsion contain mycobacterial cell wall components (not used clinically)
ISCOMs: experimental, multimeric antigen.
IL2

191
Q

Which medications are used in the treatment of anaphylaxis and at what doses?

A
IM adrenaline (0.5 mg) 
IV chlorpheniramine (10 mg) 
IV hydrocortisone (200 mg) 

Also give IV fluids, oxygen and nebulised bronchodilators

192
Q

What are the two types of latex allergy?

A

Type 1 hypersensitivity: acute onset of classic allergic symptoms
Type 4 hypersensitivity: causes contact dermatitis which arises 24-48 hours after exposure, itchy rash, NOT responsive to antihistamines

193
Q

What are SPUR infection?

A

Serious
Persistent
Unusual
Recurrent

194
Q

Describe the mechanism of action of serum sickness.

A

Penicillin binds to cell surface proteins
This acts as a neo-antigen, which stimulates a very strong IgG response
This means that the individual is sensitised to penicillin
Subsequent exposure leads to the formation of immune complexes with the penicillin and the production of more IgG antibodies
Immune complexes deposit in joints, kidneys and skin causing arthralgia, glomerulonephritis and a vasculitic rash

195
Q

How can serum sickness due to penicillin be investigated?

A

Low serum C3 + C4 (suggests classical pathway activation)
Specific IgG to penicillin
Biopsy of skin or kidneys (showing infiltration of macrophages and neutrophils, deposition of IgG, IgM and complement)

TREATMENT: stop penicillin, give steroids

196
Q

Explain the following clinical manifestations of serum sickness:

a. Deterioration in renal function
b. Disorientation
c. Purpura

A

a. Deterioration in renal function
Deposition of immune complexes in the glomeruli leads to complement activation and inflammatory cell recruitment
This leads to glomerulonephritis resulting in a rise in creatinine, proteinuria and haematuria
b. Disorientation
Small vessel vasculitis affecting the cerebral vessels can compromise the oxygen supply to the brain
c. Purpura
Inflamed blood vessels are likely to leak resulting in local haemorrhage

197
Q

What is the role of PTPN22 and which polymorphism is associated with rheumatoid arthritis?

A

It is a lymphocyte-specific tyrosine kinase that suppresses T cell activation
1858T allele increase susceptibility to rheumatoid arthritis, SLE and T1DM

198
Q

List some medications that are used in the treatment of rheumatoid arthritis.

A
Methotrexate 
TNF-alpha antagonists (e.g. infliximab)
Rituximab
Abatacept 
Tocilizumab
199
Q

Which antibody is seen in mixed connective tissue disease?

A

Anti-U1RNP

200
Q

Which antibody is present in paraneoplastic cerebellar degeneration?

A

Anti-purkinje cell antibody

201
Q

How is myeloperoxidase deficiency distinguished from chronic granulomatous disease?

A

Myeloperoxidase deficiency has a normal NBT test and patients are generally asymptomatic, however, they can develop invasive Candida infections
Chronic granulomatous disease would cause a negative NBT test (inability to generate reactive oxygen species)

202
Q

What does RANK stand for?

A

Receptor activator of nuclear factor kappa B

203
Q

Describe the presenting features of hereditary angioedema.

A

Autosomal dominant
Affect upper airway, subcutaneous tissues and abdominal organs (e.g. lip swelling, abdominal pain, diarrhoea)
Can be triggered by minor injury (e.g. dental work)

204
Q

How is allergic rhinitis treated?

A

Intranasal anti-histamine (H1) therapy - if predominant symptom is nasal discharge
Intranasal corticosteroids - if predominant symptom is nasal blockage

205
Q

Which immune modulating agents are associated with PML?

A

Natalizumab
Efalizumab
Rituximab
Mycophenolate mofetil

206
Q

What are the three types of autoimmune polyendocrinopathy syndromes?

A

Type 1: APECED
Type 2: Schmidt syndrome
Type 3: IPEX

Schmidt: Addison’s disease and hypothyroidism OR T1DM

207
Q

Name three autoimmune diseases that target type IV collagen and specify which part is targeted.

A

Goodpasture’s = non-collagenous domain of alpha-3 unit
Thin membrane disease = alpha-4 unit
Alport = alpha-5 unit

208
Q

How is oral allergy syndrome treated?

A

Oral antihistamines (provided there is no ABC compromise)

209
Q

The levels of which complement protein decreases first in active lupus?

A

C4

210
Q

What is a clinical feature of leucocyte adhesion deficiency that may present at birth?

A

Delayed separation of the umbilical cord

Also associated with severe skin infections

211
Q

List and describe three mechanisms of type IV delayed hypersensitivity.

A

Th1 –> macrophage –> TNFa (auto/alloimmunity)
Th2 –> eosinophil –> inflammatory mediators (allergy)
CTL –> granzyme and perforin –> apoptosis (auto/alloimmunity)

212
Q

Which antigen is targeted in multiple sclerosis?

A

Myelin basic protein
Proteolipid protein

NOTE: CD4 cells are mainly involved in the pathophysiology

213
Q

Which HLA genotype is associated with juvenile idiopathic arthritis?

A

DR8

214
Q

What is the receptor phenotype of Tregs?

A

CD4
CD25 (IL-2 receptor)
CTLA4
FoxP3

215
Q

List some examples of anti-TNFa agents and state some indications.

A

Examples: infliximab (IV), adalimumab, certolizumab, golimumab
Uses: rheumatoid arthritis, ankylosing spondylitis, IBD, psoriasis

216
Q

List some uses of plasmapheresis.

A

Antibody mediated disease (type II hypersensitivity)
Goodpasture’s syndrome
Myasthenia gravis
Antibody-mediated graft rejection

217
Q

Outline a common pre-transplant and post-transplant treatment regimen.

A

Pre-transplant: suppress T cells with alemtuzumab, basiliximab or anti-thymocyte globulin
Post-transplant: calcineurin inhibitor + mycophenolate with or without steroids

218
Q

Which malignancies are transplant recipients at increased risk of?

A
Viral-associated malignancy
Kaposi sarcoma
Lymphoproliferative (EBV)
Lung 
Colon
Skin (x 20)

NOTE: they also have a 20 x increased risk of MI

219
Q
Briefly describe the pathology of the following types of transplant rejection:
Hyperacute 
Acute cellular 
Acute antibody-mediated
Chronic
A

Hyperacute - thrombosis and necrosis (preformed antibodies)
Acute cellular - cellular infiltrate (CD4-mediated)
Acute antibody-mediated - vasculitis
Chronic - fibrosis, glomerulonephritis, bronchiolitis obliterans

220
Q

Which diseases may cause a false-negative on the Mantoux test?

A

HIV
Sarcoidosis
Lymphoma
Current infection

221
Q

Which vaccines are not safe in HIV?

A

Yellow fever
BCG

IMPORTANT: MMR is safe

222
Q

Which receptors does HIV use to bind and enter T cells?

A

CD4
CXCR4
CCR5

NOTE: mutation in CCR5 confers innate resistance to HIV

223
Q

Name an entry inhibitor that may be used to treat HIV.

A

Maraviroc

224
Q

What are the clinical features of Kearns-Sayre syndrome?

A
Ptosis 
Ophthalmoplegia
Pigmented retinopathy
Proximal muscle weakness 
Cardiac conduction defects 
Hearing loss and cerebellar ataxia 
Endocrine dysfunction (e.g. hypoparathyroidism, diabetes)
225
Q

What is Hirata’s disease?

A

Insulin autoimmune syndrome
Defined by fasting blood glucose and autoantibodies to serum insulin
Prevalent in Japan

226
Q

Which antigen is targeted in Guillain-Barre syndrome?

A

Ganglioside LM1

227
Q

List some features of mucosal surfaces that act as a barrier to pathogens.

A
Mucus traps pathogens 
Mucus contains secretory IgA 
Lactoferrin starves bacteria of iron 
Cilia trap pathogens and waft mucus 
Lysozyme
228
Q
Name the macrophages found in the following tissues:
Kidney
Spleen
Neural tissue
Connective tissue
A

Kidney - mesangial cell
Spleen - sinusoidal lining cell
Neural tissue - microglia
Connective tissue - histiocyte

229
Q

Which proteins can opsonise pathogens?

A

Antibodies
Complement proteins
Acute phase proteins

230
Q

What do the activating receptors on NK cells recognise?

A

Heparan sulphate proteoglycans

231
Q

Which part of the immunoglobulin molecule determines its effector function?

A

Constant region (Fc) of the heavy chain

232
Q

Which cell surface proteins are detected via the alternative complement pathway in Gram-positives and Gram-negatives?

A
Gram-positive = teichoic acid 
Gram-negative = lipopolysaccharide
233
Q

Describe the clinical features of C1q deficiency.

A

Severe childhood-onset SLE with normal levels of C3 and C4

234
Q

List some causes of basophilic stippling.

A

Beta thalassemia
Lead poisoning
Alcoholism
Sideroblastic anaemia

235
Q

Which other diseases are strongly associated with coeliac disease?

A

Dermatitis herpetiformis
Type 1 diabetes mellitus
Autoimmune thyroid disease
Down syndrome

236
Q

What is the main role of T follicular cells?

A

They help B cells undergo germinal centre reactions (thereby allowing them to become memory cells)

237
Q

In which group of antibody deficiency patients would vaccination be a worthwhile intervention?

A

IgA deficiency

Other types of antibody deficiency wont be able to mount a response to the antigen

238
Q

Describe the differences in IgG/C3 staining patterns of the three types of crescentic glomerulonephritis.

A

Immune complex: granular
Pauci immune: scanty or absent
Anti-GBM: linear

239
Q

What is the difference between immature and mature dendritic cells?

A

Immature: phagocytose small quantities of surrounding tissue
Mature: capable of migrating to lymph nodes and activating T cells

240
Q

How can hereditary angioedema be distinguished from drug-induced angioedema?

A

Hereditary angioedema: does NOT respond to antihistamines (may respond to icatibant or C1 esterase)
Drug-induced angioedema: responds to antihistamines and steroids

241
Q
Which diseases are caused by mutations in:
Mevlonate kinase (MK) gene
TNF receptor (TNFRSF1)
A
Mevalonate kinase (MK) gene - Hyper IgD with periodic fever syndrome 
TNF receptor (TNFRSF1) - TNF receptor associated periodic syndrome
242
Q

List some mutations that are associated with ankylosing spondylitis.

A

IL23 receptor - promotes differentiation of Th17
ERAP1 (ARTS1) - trims peptides for presentation on MHC
ANTXR2 - involves in maintaining structure of basement membrane
ILR2 - decoy receptor that inhibits IL1
HLA-B27 - present antigen to CD8 T cell

243
Q

What are the main targets of immunosuppression in ankylosing spondylitis?

A

TNF-alpha

IL17

244
Q

Which mediators released by mast cells are pre-formed and which are synthesised?

A

Pre-formed: histamine, serotonin, proteases
Synthesised: leukotrienes, prostaglandins, bradykinin, cytokines

NOTE: IgE binds to Fc epsilon receptors on mast cells and basophils

245
Q

Which type of hypersensitivity is most commonly implicated in autoimmune disease?

A

Type 2

246
Q

Describe the typical presentation of progressive multifocal leukoencephalopathy.

A

Clumsiness
Progressive weakness
Visual, speech, and sometimes personality changes

247
Q

Describe the immune response to allergens, worms and venoms.

A

Damaged epithelium releases signalling cytokines (e.g. TSLP)
These cytokines act on Th2, Th9 and ILC2 cells to produce IL4, IL5 and IL13
These cytokines act on eosinophils and basophils which are important in expelling parasites and allergens
The cytokines from the damages epithelium can also activate Th2 cells, which release IL4
IL4 stimulates B cells to produce IgE and IgG4

248
Q

What is the maximum number of important HLA mismatches that a organ recipient can have with a donor?

A

6

There are three important HLA types (A, B and DR) and there are 2 alleles for each

249
Q

Which cytokine is important for promoting eosinophil growth?

A

IL5

250
Q

Which interleukin is important in sensitisation to allergic stimuli?

A

IL12

251
Q

Which medications are used to treat granulomatosis with polyangiitis?

A

Inducing Remission: rituximab, cyclophosphamide, steroids
Maintaining Remission: azathioprine, methotrexate, rituximab
NOTE: plasma exchange is used in severe cases

252
Q

What could cause a raised CH50?

A

Inflammation (e.g. rheumatoid arthritis)

253
Q

Which anti-TNF agent is NOT effective in inflammatory bowel disease?

A

Etanercept (its Fc region is not functional)

254
Q

What are some therapeutic targets in psoriasis?

A

IL12/23
TNF-alpha
IL17

255
Q

What are some therapeutic targets in rheumatoid arthritis?

A

IL6
TNF-alpha
B cells

256
Q

Describe an infusion reaction that might occur with the use of biologic agents.

A

Urticaria, hypotension, tachycardia, wheeze (IgE)

Headache, myalgia, fever

257
Q

Describe injection site reactions that you might see when using biologic agents.

A

Peak reaction at 48 hours
Recall reactions - may occur at the site of previous injection
Mixed cellular infiltrate (often with CD8)
Not usually IgE or immune complex-mediated

258
Q

Which infections are patients receiving biologic therapy at increased risk of?

A
TB
HBV and HCV
HIV 
CMV 
JC Virus
259
Q

Which malignancies are patients receiving biologic therapy at risk of?

A

Lymphoma (EBV)
Non-melanoma skin cancer
Melanoma

260
Q

What do TNF and TGF stand for?

A

Tumour necrosis factor

Transforming growth factor

261
Q

Which cell line is used to test for ANA?

A

Hep-2 (human epidermoid cancer line)

262
Q

Describe the immunofluorescence pattern seen in lupus nephritis.

A

Granular lumpy bumpy pattern

263
Q

List three ANA staining patterns and the associated antibodies/diseases.

A

Homogenous - anti-dsDNA
Speckled - Ro, La, Sm, U1RNP
Nucleolar - anti-Scl70 (diffuse systemic sclerosis)
Cytoplasmic - anti-Jo1, anti-SRP (also nuclear)

NOTE: anti-centromere antibodies have a different staining pattern

264
Q

What is typically used as initial therapy in the prevention of allograft rejection?

A

Azathioprine/sirolimus + prednisolone + mycophenolate mofetil

265
Q

Which conditions are TNF-alpha inhibitors used to treat?

A
Rheumatoid arthritis 
Ankylosing spondylitis 
Psoriasis
IBD
NOTE: etanercept is ineffective in IBF
266
Q

Which conditions are ustekinumab used to treat?

A

Psoriasis

Crohn’s disease

267
Q

Which conditions are secukinumab used to treat?

A

Psoriasis

Ankylosing spondylitis

268
Q

What is abatacept used to treat?

A

Rheumatoid arthritis

269
Q

What are some consequences of deficiency in the mannose binding lectin pathway?

A

Does NOT usually cause significant immunodeficiency
May cause increased risk of infection in patients with other reasons for immune deficiency (e.g. chemotherapy, HIV, premature infants, antibody deficiency)

270
Q

How is ACEi-induced angioedema different from other types of drug-induced angioedema?

A

DOES NOT RESPOND TO ANTIHISTAMINES
Slower progression than histamine-mediated angioedema (over hours)
No urticaria or itching