Immunology Flashcards

1
Q

skin - barriers to infection

A
  • consists of tightly packed keratinised cells
  • this physically limits colonisation by microorganisms
  • low pH
  • low oxygen tension
  • sebaceous glands: hydrophobic oils that repel water & microorganisms, lysozymes that destroy the structural integrity of the bacterial cell wall, ammonia & defensins that have antibacterial properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

mucosal surfaces - barriers to infection

A
  • mucus is a physical barrier that traps invading pathogens
  • contains secretory IgA which binds to pathogens and prevents bacteria and viruses from attaching to and penetrating epithelial cells
  • lysozyme and antimicrobial peptides directly kill invading pathogens
  • lactoferrin starves invading bacteria of iron
  • cilia directly trap pathogens and contribute to the removal of mucous, which is assisted by physical manoeuvres such as sneezing and coughing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

benefits of commensal bacteria

A
  • they compete with pathogenic bacteria for scarce resources
  • they produce fatty acids and bactericidins that inhibit the growth of many pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

components of the innate immune system

A

cells
- polymorphonuclear cells (neutrophils, eosinophils, basophils)
- monocytes and macrophages
- NK cells
- dendritic cells

soluble components
- complement
- acute phase proteins
- cytokines and chemokines

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

key features of cells of the innate immune system

A
  • essentially identical responses in ALL individuals
  • cells express receptors that allow them to detect and home to sites of infection
  • cells express genetically encoded receptors (PRRs) that allow them to detect pathogens at the site of infection
  • cell has phagocytic capacity that allows them to engulf pathogens
  • cells secrete cytokines and chemokines that regulate the immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

polymorphonuclear cells

A
  • produced in bone marrow
  • migrate rapidly to site of injury
  • express receptors for cytokines/chemokines - to detect inflammation
  • express pattern recognition receptors - to detect pathogens
  • express Fc receptors for Ig - to detect immune complexes
  • capable of phagocytosis/oxidative & non-oxidative killing - particularly neutrophils
  • release enzymes, histamine, lipid mediators of inflammation from granules
  • secrete cytokines and chemokines to regulate inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

relationship between monocytes and macrophages

A

monocytes are produced in the bone marrow, circulate in the blood and migrate to tissues where they differentiate into macrophages

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

macrophages

A
  • present within tissue
  • express receptors for cytokines and chemokines - to detect inflammation
  • express pattern recognition receptors - to detect pathogens
  • express Fc receptors for Ig - to detect immune complexes
  • capable of phagocytosis / oxidative and non-oxidative killing
  • secrete cytokines and chemokines to regulate inflammation
  • capable of presenting processed antigen to T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

how are macrophages different to polymorphonuclear cells?

A

they can process antigens and present it to T cells

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

phagocyte recruitment

A
  • cellular damage and bacterial products trigger the local production of inflammatory mediators (cytokines and chemokines)
  • cytokines will activate the vascular endothelium enhancing permeability
  • chemokines attract phagocytes (NB: macrophages are already present at peripheral sites)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

recognition of microorganisms

A
  • pattern recognition receptors (PRRs) like Toll-like receptors and mannose receptors recognise generic motifs known as pathogen-associated molecular patterns (PAMPs) such as bacterial sugars, DNA and RNA
  • Fc receptors on these cells allows them to bind to the Fc portion of immunoglobulins thereby allowing the phagocytes to recognise immune complexes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

what is opsonisation? what does it facilitate?

A

it facilitates endocytosis
- opsonins act as a bridge between the pathogen and the phagocyte receptor
- the antibodies will bind to Fc receptors on the phagocytes
- complement components can bind to complement receptors (e.g., CR1)
- acute phase proteins (e.g, CRP) will also promote phagocytosis

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

formation of phagolysosome

A
  • the pathogen is taken up into a phagosome
  • the phagosome will fuse with a lysosome to form a phagolysosome
  • this is procted comparment in which killing of the organism occurs
  • the killing of the pathogen can occur by oxidative mechanisms or non-oxidative mechanisms
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

non-oxidative killing within the phagolysosome

A
  • killing by the release of bacteriocidal enzymes such as lactoferrin and lysozyme into the phagolysosome
  • enzymes are present in granules
  • each has a unique antimicrobial spectrum
  • this results in a broad range of cover against bacteria and fungi
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

death of the phagocyte - neutrophils

A
  • the process of phagocytosis depleted glycogen reserves within the neutrophil, which is then followed by neutrophil death
  • as the cells die, residual enzymes are released which causes liquefaction of the adjacent tissue
  • accumulation of dead and dying neutrophils within the infected tissue results in the formation of pus
  • an accumulation of pus forms an abscess
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

summary of innate response to infection

A
  • expression of endothelial activation markers
  • mobilisation of phagocytes and precursors from bone marrow or within tissues
  • increased neutrophil adhesion and migration into tissues
  • macrophage - T cell communication
  • phagocytosis of organisms
  • oxidative and non-oxidative killing
  • cell death and formation of pus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

natural killer cells

A
  • these are a type of lymphocyte
  • present within the blood and may migrate to inflamed tissue
  • express inhibitory receptors for self HLA which prevents inappropriate activation by normal self cells
  • express a range of activatory receptors including natural cytotoxicity receptors that recognise heparan sulphate proteoglycans
  • integrate signals from inhibitory and activatory receptors (usually the inhibitory signals will dominate)
  • they are cytotoxic - kill ‘altered self’ cells (i.e., malignancy or virus-infected cells)
  • secrete cytokines to regulate inflammation and promote dendritic cell function
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

dendritic cells

A
  • reside in peripheral tissues
  • express receptors for cytokines and chemokines (to detect inflammation)
  • express pathogen recognition receptors (to detect pathogens)
  • express Fc receptors for immunoglobulin (to detect immune complexes)
  • capable of phagocytosis
    following phagocytosis, dendritic cells will:
    a. upregulate expression of HLA molecules
    b. express co-stimulatory molecules
    c. migrate via lymphatics to lymph nodes (mediated by CCR7)
  • present processed antigen to T cells in lymph nodes to prime the adaptive immune system
  • express cytokines to regulate the immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

adaptive immune system: components

A

humoral immunity
- B lymphocytes and antibody

cellular immunity
- T lymphocytes (CD4+ and CD8+)

soluble components
- cytokines and chemokines

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

what are primary lymphoid organs?

A

= organs involved in lymphocyte development

Bone marrow
- both T and B lymphocytes are derived from haematopoietic stem cells in the bone marrow
- it is the site of B cell maturation

Thymus
- site of T cell maturation
- most activate in the foetal and neonatal period, involutes after puberty

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

what are secondary lymphoid organs?

A

= anatomical sites of interaction between naive lymphocytes and microorganisms

examples:
- spleen
- lymph nodes
- MALT

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

key features of cells of the adaptive immune response

A
  • wide repertoire of antigen receptors
  • exquisite specificity
  • clonal expansion
  • immunological memory
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

T lymphocyte maturation

A
  • arise from haematopoietic stem cells in the bone marrow
  • exported as immature cells to the thymus where undergo selection
  • mature T lymphocytes enter the circulation and reside in secondary lymphoid organs
  • they undergo positive and negative selection before being exported to the periphery
  • CD4+ recognises peptides presented by HLA class II
  • CD8+ recognises peptides presented by HLA class I
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

CD4+ T cells (Helper T cells)

A
  • recognises peptides derived from extracellular proteins
  • these peptides are usually presented on HLA Class II molecules (HLA-DR, HLA-DP, HLA-DQ)
  • they have important immunoregulatory functions of a full B cell response:
    a. they provide help for the development of a full B cell response
    b. they provide help for the development of some CD8+ T cell responses
  • different cytokines promote development along different lines of development
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

CD8+ cytotoxic T cells

A
  • specialised cytotoxic cells
  • recognise peptides derived from intracellular proteins presented on HLA class I (HLA-A, HLA-B and HLA-C)
  • kills cells directly:
    a. perforin (pore-forming) and granzymes
    b. expression of Fas ligand
  • secrete cytokines (e.g., IFN-gamma, TNF-alpha)
  • particularly important in defence against viral infections and tumours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

T cell memory

A
  • response to successive exposures of antigen is qualitatively and quantitatively different from that of first exposure
  • there is a pool of memory T cells that are ready to respond to antigen
  • these cells are more easily activated than naive cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

B lymphocyte maturation

A

they initially exist in the periphery as IgM B cells, but they can undergo a germinal centre reaction to differentiate into plasma cells expressing IgG, IgE and IgA

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

central tolerance

A

if the receptors on the B cells in the bone marrow recognise self, they will be deleted

if not, they will survive

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

antigen encounter by B cells - early IgM response

A

if the B cell in the periphery engages an antigen you can get an early IgM response where the cell differentiates into an IgM secreting plasma cell

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

antigen encounter by B cells - germinal centre reaction

A
  • this is dependent on T helper cells
  • firstly, dendritic cells will prime the CD4+ T helper cells
  • the CD4+ cells then provide help for B cell differentiation
  • this interaction is mediated by CD40L:CD40
  • with the help of CD4+ cells, the B cells will proliferate
  • they then undergo somatic hypermutation and isotype switching (from IgM to IgG/A/E)
  • they will then become plasma cells which produce antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

what are immunoglobulins?

A
  • soluble proteins made up of 2 heavy chains and 2 light chains
  • heavy chain determines the antibody class (GAMED)
  • they are subclasses of IgG and IgA
  • antigen is recognised by the antigen binding region (Fab) which is made up of both the heavy and light chains
  • effector function is determined by the constant region (Fc) of the heavy chain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

function of antibodies

A

identification of pathogens and toxins (Fab-mediated)

interact with other components of immune responses to remove pathogens (Fc-mediated):
- complement
- phagocytes
- NK cells
particularly important in defence against BACTERIA of all kinds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

what is complement?

A

20+ tightly regulated, linked proteins
- produced by the liver
- present in the circulation as inactive molecules

when triggered, they will enzymatically activate other proteins in a biological cascade
- results in a rapid, highly amplified response

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

which are the 3 pathways of complement activation?

A
  1. classical
  2. mannose binding lectin pathway
  3. alternative pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

complement activation - classical pathway

A
  • activated by immune complexes
  • the formation of antibody-antigen immune complexes results in a conformational change in antibody shape which exposes the binding site for C1
  • binding of C1 to the antibody results in activation of the cascade
  • as it involves antibodies, it does depend on the activation of the adaptive immune response (i.e., it will NOT occur very early in the immune response)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

complement activation - mannose binding lectin (MBL) pathway

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)
  • this is NOT dependent on the adaptive immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

complement activation - alternative pathway

A

this is directly triggered by binding of C3 to bacterial cell wall components
e.g., lipopolysaccharide of gram-negative bacteria
e.g., teichoic acid of gram-positive bacteria
- this is NOT dependent on the acquired immune response
- involves factors: B, I, P

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

complement activation: what is the membrane attack complex? how is it activated and what is its role?

A
  • activation of C3 convertase is the major amplification step
  • this triggers the formation of the membrane attack complex via C5-9
  • the membrane attack complex makes holes in membranes
  • complement fragments that are released during complement activation play various roles in the immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

cytokines

A
  • small protein messengers
  • immunomodulatory function
  • autocrine and paracrine dependent action
  • examples: IL2, IL6, IL10, IL12, TNF-alpha, TGF-beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

chemokines

A
  • subset of cytokines
  • direct recruitment/homing of leukocytes in an inflammatory response
  • CCL19 and CCL21 are ligands for CCR7 and important in directing dendritic cell trafficking to lymph nodes
  • other examples: IL8, RANTES, MIP-1 alpha and beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

classification of immunodeficiencies

A

primary (inherited)
- very rare
- >100 types of primary immunodeficiency

secondary (acquired)
- common
- often subtle
- often involves more than 1 component of the immune system

physiological (expected)
- neonates
- elderly
- pregnancy

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

examples of secondary immunodeficiency

A

infection:
- HIV
- measles virus
- mycobacterial infection

biochemical disorders:
- malnutrition
- specific mineral deficiencies (zinc, iron)
- renal impairment

malignancy:
- myeloma
- leukaemia
- lymphoma

drugs:
- corticosteroids
- anti-proliferative immunosuppressants
- cytotoxic agents

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

clinical features of immunodeficiencies

A

infections:
- 2 major OR 1 major + recurrent minor infections in 1 year
- unusual organisms
- unusual sites
- unresponsive to treatment
- chronic infections
- early structural damage

features suggestive of primary immunodef:
- family history
- young age at presentation
- failure to thrive

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

components of the innate immune system

A

cells
- polymorphonuclear cells (neutrophils, eosinophils, basophils): produced in the bone marrow and migrate rapidly to the site of injury
- monocytes and macrophages: monocytes are produced in the bone marrow, circulate in blood and migrate to tissues where they differentiate to macrophages
- dendritic cells
- NK cells

soluble components
- complement
- acute phase proteins
- cytokines and chemokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

phagocytes

A
  • do NOT tend to vary much between individuals
  • express cytokine/chemokine receptors that allow them to home into sites of infection
  • cells have pattern recognition receptors (e.g., Toll-like receptors) which recognise generic motifs known as pathogen-associated molecular patterns (PAMPs) such as bacterial sugars, DNA and RNA
  • cells have Fc receptors to allow the detection of immune complexes
  • they also have phagocytic capacity meaning that they can engulf the pathogens
  • cells can secrete cytokines and chemokines to regulate the immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

overview of response to infection

A
  • the presence of an infection in the tissues will stimulate endothelial activation and the expression of adhesion molecules
  • neutrophils will mobilise from the bone marrow and enter the blood stream
  • they will adhere to the endothelium at the site of damage and migrate into the tissue
  • tissue resident macrophages will phagocytose the pathogens
  • macrophages will process the antigens and present them to T cells
  • neutrophils eventually die and form pus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

conditions that cause a failure to produce neutrophils

A

failure of stem cells to differentiate along myeloid or lymphoid lineage: RETICULAR DYSGENESIS
- autosomal recessive SCID
- you get no lymphoid or myeloid cells

specific failure of neutrophil maturation:
KOSTMANN SYNDROME
- autosomal recessive severe congenital neutropaenia
CYCLIC NEUTROPAENIA
- autosomal dominant episodic neutropaenia

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

condition that causes a defect of phagocyte migration

A

Leukocyte adhesion deficiency
- deficiency of CD18
- during an infection, neutrophils will be mobilised from the bone marrow however they will NOT be able to access the site of infection in the tissues
- high neutrophil count in the blood, NO pus formation

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

condition that causes a failure of oxidative killing mechanisms

A

chronic granulomatous disease
- absent respiratory burst (inability to generate oxygen free radicals)
- excessive inflammation
- granuloma formation
- lymphadenopathy and hepatosplenomegaly

cytokine deficiency
- there is a cytokine cycle between macrophages and T cells
- patients vulnerable to organisms that infect macrophages
- most of these patients present with atypical mycobacterial infections (and sometimes salmonella)

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

how do we investigate chronic granulomatous disease?

A

2 tests

Nitroblue Tetrazolium (NBT)
- changes from yellow to blue

Dihydrorhodamine (DHR)
- becomes fluorescent

  • normally, when neutrophils are activated, a respiratory burst takes place and hydrogen peroxide is produced
  • both of these tests are looked at the ability of neutrophils to produce hydrogen peroxide and generate oxidative stress
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

treatment of phagocyte deficiencies

A

aggressive management of infection
infection prophylaxis:
- antibiotics (e.g., Septrin)
- anti-fungals (e.g., itraconazole)

definitive therapy
- haematopoietic stem cell transplantation
- specific treatment for chronic granulomatous disease: IFN-gamma therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

what type of infections do NK cell deficiencies increas the risk of?

A

VIRAL
- herpes simplex
- VZV
- EBV
- CMV
- HPV

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

treatment of NK cell deficiencies

A
  • prophylactic antiviral drugs (e.g., aciclovir, ganciclovir)
  • cytokines to stimulate NK cytotoxic function
  • haematopoietic stem cell transplantation (if severe)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

phenotype of Kostmann syndrome

A

recurrent infections with NO neutrophils on FBC

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

phenotype of leukocyte adhesion deficiency

A

recurrent infections with HIGH neutrophil count but no abscess formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

phenotype of chronic granulomatous disease

A

infection with atypical mycobacterium
normal FBC

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

phenotype of classical NK cell deficiency

A

severe chickenpox, disseminated CMV infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

deficiency of complement

A
  • may involve the classica, alternate, C3 or final common pathway
  • increases susceptibility to bacterial infections
  • particularly increases susceptibility to encapsulated bacterial infections (Neisseria meningitides, Haemophilus influenzae, Streptococcus pneumoniae)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

clinical phenotype of complement deficiency

A
  • almost ALL patients with C2 deficiency have SLE
  • they usually have severe skin disease
  • they also have an increased risk of infections
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

functional complement tests

A

CH50 is a test of the classical pathway
- testing the activity of C1, 2 and 4, C3 and C5-9

AP50 is the test of the alternate pathway
- testing the activity of B, D, properidin, C3 and C5-9

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

summary of investigating complement

A
  • C1q deficiency is an inherited form of complement deficiency that tends to present with SLE in childhood
  • in patients with C1q deficiency, they will not be able to activate their classical pathway so CH50 will be low
  • with C9 or C7 deficiency, both the CH50 and AP50 will be low because the problem lies in the common pathway
  • if someone has acquired SLE, they will have low C4 and possibly low C3
  • they may also have some dysfunction of the CH50 pathway because they don’t have much complement left
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

management of complement deficiencies

A

vaccination
- boost protection mediated by other arms of the immune system
- they should be vaccinated against polysaccharide encapsulated bacteria
- e.g., meningovax, pneumocax, HIB vaccines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

phenotype of C1q deficiency

A

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

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

phenotype of C3 deficiency with presence of a nephritic factor

A

membranoproliferative nephritis and abnormal fat distribution

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

phenotype of C7 deficiency

A

meningococcus meningitis with family history of sibling dying of the same condition aged 6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

MBL deficiency phenotype

A

recurrent infections when neutropaenic following chemotherapy but previously well

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

summary of types of primary immunodeficiency

A

phagocytes
- Kostmann syndrome
- leukocyte adhesion deficiency
- chronic granulomatous disease

natural killer cells
- classical NK deficiency
- functional NK deficiency

complement
- classical pathway deficiencies
- alternative pathway deficiencies
- C3 deficiency
- terminal pathway deficiencies

haematopoietic stem cells
- reticular dysgenesis

cytokines
- IL12 and IL12 receptor deficiency
- IFNgamma and IFNgamma receptor deficiency

lymphoid precursors
- severe combined immunodeficiency

T cells
- 22q11.2 deletion syndrome
- bare lymphocyte syndrome

B cells
- X-linked agammaglobulinaemia
- X-linked hyperIgM syndrome
- common variable immunodeficiency
- IgA deficiency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

causes of microcytic anaemia

A
  • iron deficiency
  • thalassaemia trait
  • anaemia of chronic disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

is anisopoikilocytosis (in the context of microcytic anaemia) more associated with iron deficiency or thalassaemia trait?

A

iron deficiency

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

causes of aggregated ribosomal material on blood film

A
  • beta-thalassaemia trait
  • lead poisoning
  • alcoholism
  • sideroblastic anaemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

hypersegmented neutrophils - feature of which type of anaemia

A

megaloblastic
(reflected impaired DNA synthesis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

causes of megaloblastic anaemia

A
  • B12 deficiency
  • folate deficiency
  • drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

causes of target cells

A
  • iron deficiency
  • thalassaemia
  • hyposplenism
  • liver disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

what are Howell-Jolly bodies? cause?

A

nuclear remnants visible within RBCs
- hyposplenism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

causes of iron deficiency

A
  • blood loss
  • poor diet
  • malabsorption
  • combinations of the above
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

causes of megaloblastic change

A

B12/folate deficiency:
- poor diet
- malabsorption
- pernicious anaemia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

causes of hyposplenism

A

absent spleen
- therapeutic
- trauma

poorly-functioning spleen
- IBD
- coeliac disease
- SCD
- SLE

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

deficiency seen in coeliac disease

A

iron, B12, folate, fat, calcium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

deficiency seen in crohn’s

A

B12, bile salts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

deficiency seen in pancreatic disease

A

fat, calcium, B12

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

deficiency seen in infective/post-infective disease

A

fat, folate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

gold standard investigation for coeliac disease

A

upper GI endoscopy and distal duodenal biopsies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

which HLA mutation is most commonly associated with coeliac

A

HLA DQ2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

coeliac disease: T-cell response to gluten

A
  • peptides from gluten (i.e., gliadin) are deamidated by tissue transglutaminase
  • the deamidated gluten is taken up by antigen-presenting cells
  • it is then presented by HLA molecules to CD4+ T cells
  • CD4+ T cell activation results in secretion of IFN-gamma and may directly lead to increased IL-15 secretion
  • the cytokines promote activation of the intra-epithelial lymphocytes
  • these T-cells are gamma-delta T cells
  • these intra-epithelial lymphocytes kill epithelial cells via the NKG2D receptor
  • in other words, the intraepithelial lymphocytes cause damage to the gut wall and lead to the presentation of coeliac disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

coeliac disease: B cell response to gluten

A
  • there may be B cells whose antibodies recognise gliadin
  • the B cells will process the antigens and present them via the HLA molecule to the CD4+ T cell
  • these primed T cells will then be able to provide help to the B cells that are trying to undergo germinal centre reactions
  • the B cells will then undergo isotype switching and affinity maturation to become memory cells and plasma cells which will produce antibodies that are specific for gliadin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

serological tests for coeliac disease

A

IgA anti-transglutaminase antibody
IgA anti-endomysial antibody

NB: both anti-TTG and anti-endomysial antibody assays routinely measure IgA antibodies, and so these are not useful in IgA deficient patients. 1 in 600 are IgA deficient.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

coeliac histology: key features

A
  • villous atrophy with crypt hyperplasia
  • intra-epithelial lymphocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

conditions other than coeliac that are associated with increased intraepithelial lymphocytes

A
  • dermatitis herpetiformis
  • giardiasis
  • cows milk protein sensitivity
  • IgA deficiency
  • tropical sprue
  • post-infective malabsorption
  • drugs (NSAIDs)
  • lymphoma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

complications of coeliac

A
  • malabsorption
  • osteomalacia and osteoporosis
  • neurological disease (epilepsy, cerebral calcification)
  • lymphoma: multi-focal T cell lymphoma, very difficult to treat
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

consequences of undiagnosed coeliac disease

A

deficiencies:
- iron deficiency
- folate/B12 deficiency
- vit D and vit K deficiency

  • dietary compliance protects against malignancy
  • often feel better physically and psychologically
  • NB: malabsorption can lead to reduced absorption of medications (e.g., thyroid)
  • mortality of untreated: 2-3x general population (lymphoma, infection)
  • mortality returns to normal after gluten-free for 3-5 years
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

components of the adaptive immune system

A

T lymphocytes
- CD4 T cells
- CD8 T cells

B lymphocytes
- B cell
- plasma cells
- antibodies

soluble components
- cytokines and chemokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

lymphoid development

A
  • lymphocytes are derived from stem cells in the BM
  • some will progress along the T cell lineage - they will leave the BM and undergo thymic selection before exiting as mature CD4 and CD8 T cells
  • on the other hand, the cells can leave the bone marrow as Pro B cells or Pre B cells which then become IgM B cells
  • these can undergo germinal centre reactions and mature into memory B cells and plasma cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

what is reticular dysgenesis?

A
  • most severe form of severe combined immunodeficiency (SCID)
  • mutation in mitochondrial energy metabolism enzyme adenylate kinase 2 (AK2)

Failure of production of:
- lymphocytes
- neutrophils
- monocyte/macrophages
- platelets

fatal in very early life unless corrected with BM transplantation

NB: it is called severe combined because it affects both B and T lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

X-linked SCID

A
  • most common form of SCID (45%)
    Caused by mutation of common gamma chain on chromosome Xq13.1:
  • this is a component of many cytokine receptors including IL2, IL4, IL7, IL9, IL15, IL21
  • the inability to respond to cytokines causes early arrest of T cell and NK cell development and the production of immature B cells

Phenotype:
- very low or absent T cell numbers
- normal or increased B cell numbers (but low immunoglonulin)
- very low or absent NK cell numbers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

ADA deficiency

A
  • 16.5% of all SCID
  • caused by adenosyne deaminase deficiency
  • ezyme required by lymphocytes for cell metabolism
  • failure of maturation along any of the lineages

Phenotype:
- very low/absent T cell numbers
- very low/absent B cell numbers
- vely low/absent NK cell numbers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

clinical phenotype of SCID

A
  • unwell by 3 months of age; for the first 3 months they are protected by maternal IgG
  • infections of all types
  • failure to thrive
  • persistent diarrhoea
  • unusual skin disease
  • Fx of early death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

T lymphocyte maturation

A
  • arise from haematopoietic stem cells
  • exported as immature cells to the thymus where they undergo selection
  • CD8+ T cells recognise peptides presented by HLA class I molecules
  • CD4+ T cells recognise peptides presented by HLA class II molecules
  • mature T lymphocytes enter the circulation and reside in secondary lymphoid organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

selection and central tolerance - T cells

A
  • if the T cell has very low affinity for HLA then it is useless and so will not be selected
  • if they have very high affinity then they are dangerous because they can give rise to autoreactivity - so these are also negatively selected and die
  • so, you are left with the T cells that have intermediate affinity for HLA (about 10%)
  • they will then differentiate further depending on which type of HLA molecule they show intermediate affinity to
  • intermediate affinity for HLA class I; differentiate as CD8+ T cell
  • intermediate affinity for HLA class II; differentiate as CD4+ T cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

CD8+ cytotoxic T cells

A
  • specialised cytotoxic cells
  • recognise peptides derived from intracellular proteins in association with HLA class I (HLA-A, HLA-B, HLA-C)
  • kills cell directly
    a) perforin and granzymes
    b) expression of Fas ligand (which binds to Fas and induces apoptosis)
  • secrete cytokines
  • particularly important in defence against viral infections and tumours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

CD4+ Helper T cells

A

recognise:
- peptides derived from extracellular proteins
- presented on HLA class II molecules (HLA-DR, HLA-DP, HLA-DQ)

immunoregulatory functions via cell:cell interactions and expression of cytokines
- provide help for development of a full B cell response
- provide help for development of some CD8+ T cell responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

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

A

the thymus does NOT fully develop.
developmental defect of the pharyngeal pouch.

Facial abnormalities:
- high forehead
- low set, abnormally folded ears
- cleft palate
- small mouth and jaw

other features:
- underdeveloped parathyroid gland (resulting in hypocalcaemia)
- oesophageal atresia
- underdeveloped thymus
- complex congenital heart disease

consequences of underdeveloped thymus:
- normal B cells
- low T cell numbers
- homeostatic proliferation with age: if T cells are in an empty compartment they will just keep proliferating, so, in the end, T cell numbers tend to increase with age
- immune function is mildly impaired and tends to improve with age

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

clinical features of T lymphocyte deficiencies

A
  • viral infections (e.g., CMV)
  • fungal infections (e.g., PCP, cyptosporidum)
  • some bacterial infections, esp. intracellular organisms (TB, salmonella)
  • early malignancy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

investigation of T cell deficiencies

A
  • total white cell count and differential (NB: lymphocyte count is much higher in children)
  • lymphocyte subsets (quantify CD4, CD8, B cell, NK cell)
  • immunoglobulins (if CD4 T cell deficient, they will have IgM but no IgG or IgA)
  • functional tests of T cell activation and proliferation
  • HIV test
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

management of immunodeficiency involving T cells

A
  • aggressive prophylaxis/treatment of infection
  • haematopoietic stem cell transplantation (to replace abnormal populations in SCID, to replace abnormal cells)
  • enzyme replacement therapy (PEG-ADA for ADA-SCID)
  • gene therapy (stem cells are treated ex vivo with viral vectors containing missing components. transducent cells have a survival advantage in vivo)
  • thymic transplantation (to promote T cell development in Di George syndrome)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

central tolerance - B cells

A

no recognition of self in bone marrow - survive

recognition of self in bone marrow - negative selection to avoid autoreactivity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

B cell antigen encounter

A
  • when B cells encounter antigens, the early response tends to be an IgM response (not T cell dependent)
  • they will differentiate to form IgM memory cells and plasma cells
  • they also undergo a T cell dependent germinal centre reaction
  • dendritic cells will prime the CD4+ T cells
  • then, these CD4+ T cells help with B cell differentiation (this interaction requires CD40 ligand expression on T cells, and CD40 expression on B cells)
  • once they have received this help from CD4+ T cells, the B cells can proliferate and undergo somatic hypermutation (to refine their receptor to develop high affinity) and they can isotype switch (to IgA, IgG and IgE)
  • this results in the production of much higher affinity memory cells and plasma cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

immunoglobulins

A
  • soluble proteins made up of 2 heavy chains and 2 light chains
  • the heavy chain determines the antibody class
  • there are also subclasses of IgG and IgA
  • antigen is recognised by the antigen binding region (Fab) of both the heavy and light chains
  • effector function is determined by the constant region of the heavy chain (Fc)

Antibody function:
1) identification of pathogens and toxins (Fab-mediated)
- particularly important against extracellular pathogens
2) interacts with other components of the immune response to remove pathogens (Fc-mediated)
- complement
- phagocytes
- NK cells
3) important in defence against bacteria of all kinds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Bruton’s X-linked Hypogammaglobulinaemia

A
  • this affects B cells at the point at which they emerge from the bone marrow
  • in this condition, you do NOT have B cells maturing and, as such, you do NOT have any antibodies being produced
  • caused by an abnormal B cell tyrosine kinase (BTK) gene
  • this means that pre-B cells cannot develop into mature B cells
  • therefore, there is an absence of mature B cells
  • once maternal IgG is out of the system (around 3 months), they will have NO ANTIBODIES

clinical phenotype of Bruton’s X-linked Hypogammaglobulinaemia:
- boys present in the first few years of life
- recurrent bacterial infections
- viral, fungal, parasitic infections
- failure to thrive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

hyper IgM syndrome

A
  • this condition blocks the maturation of IgM B cells through germinal centres into B cells that produce other classes of immunoglobulins
  • the IgM B cells are unable to enter a germinal centre reaction
  • this is due to a mutation in the CD40 ligand gene
  • so, this is technically a T cell problem
  • CD4+ T cells cannot signal to B cells and help them during the germinal centre reaction

consequences:
- normal number of circulating B cells
- normal number of T cells
- NO germinal centre development with lymph nodes and spleen
- failure of isotype switching
- elevated serum IgM
- undetectable serum IgG, IgA and IgE

clinical phenotype:
- boys present in first few years of life
- recurrent infections (bacterial)
- subtle abnormality in T cell function predisposes to pneumocystitis jirovecii infection, autoimmune disease and malignancy
- failure to thrive

110
Q

common variable immunodeficiency

A

cause is unknown
can present in adolescents/adults
heterogeneous group of disorders
some form of failure of differentiation/function of B lymphocytes

defined by:
- marked reduction in IgG, with low IgA or IgM
- poor/absent response to immunisation
- absence of other defined immunodeficiency

clinical phenotype:
- may be adults or children
- recurrent bacterial infections
- pulmonary disease (obstructive airways disease, interstitial lung disease, granulomatous interstitial lung disease)
- gastrointestinal disease (IBD-like, sprue like illness, bacterial overgrowth)
- autoimmune disease (AIHA, RA, pernicious anaemia, thyroiditis, vitiligo)
- malignancy (non-Hodgkin’s lymphoma)

111
Q

selective IgA deficiency

A
  • relatively common
  • a lot of people will not be aware of it
  • 2/3rd individuals asymptomatic
  • 1/3rd have recurrent respiratory tract infections
  • genetic component, but cause is yet unknown
112
Q

summary of B cell maturation defects

A

failure of lymphocyte precursors: severe combined immunodeficiency

failure of B cell maturation: Bruton’s X-linked agammaglobulinaemia

failure of T cell costimulation: X-linked hyper IgM syndrome

failure of production of IgG antibodies: common variable immune deficiency, selective antibody deficiency

failure of IgA production: selective IgA deficiency

113
Q

clinical features of antibody deficiency (or CD4+ T cell deficiency)

A
  • bacterial infections (e.g., staphylococcus, streptococcus)
  • toxins (e.g., tetanus, diphtheria)
  • some viral infections (e.g., enterovirus)
114
Q

investigation of B cell deficiencies

A

total white count and differential
lymphocyte subsets
serum immunoglobulins and protein electrophoresis: production of IgG is a surrogate marker for CD4+ T cell helper function
functional tests of B cell function:
- specific antibody responses to known pathogens
- measure IgG antibodies against tetanus, H. influenzae B and S. pneumoniae
- if the specific antibody levels are LOW, immunise with the appropriate killed vaccine and repeat antibody measurement 6-8 weeks later
- functional tests have generally syperseded IgG subclass quantification

115
Q

protein electrophoresis in antibody deficiency

A
  • when you run your proteins through the gel you will get a big peak for albumin
  • then you will also get alpha-1, alpha-2 and beta peaks which are showing the presence of various other protens
  • at the end, you get the gamma peak - this contains all the antibodies
  • a patient who is antibody deficient will have no gamma peak
116
Q

management of immunodeficiency involving B cells

A
  • aggressive prophylaxis/treatment of infection
  • immunoglobulin replacement if required
    [derived from pooled plasma from thousands of donors, contains IgG antibodies to a variety of common organisms, aim to maintain through IgG levels within the normal range, treatment is lifelong]
  • immunisation is not worthwile because they will not be able to produce antibodies unless it is a selective IgA deficiency
117
Q

common variable immunodeficiency - typical phenotype

A

adult with bronchiectasis, recurrent sinusitis and development of atypical SLE

118
Q

X-linked hyper IgM syndrome due to CD40L mutation - typical phenotype

A

recurrent bacterial infections as a child, episode of PCP, high IgM, absent IgA and absent IgG

119
Q

Bruton’s X-linked hypogammaglobulinaemia - typical phenotype

A

1 year old boy, recurrent bacterial infections, CD4 and CD8 T cell present, B cell absent, IgG IgA and IgM absent

120
Q

IgA deficiency - typical phenotype

A

recurrent respiratory tract infections, absent IgA, normal IgM and IgG

121
Q

what is immunopathology?

A
  • it is damage to the host caused by the immune response
  • quite often the problems that occur after infection will actually be a result of the immune response rather than the pathogen itself (e.g., abscesses)
122
Q

auto-inflammatory vs autoimmune disease

A

autoinflammatory or autoimmune disease refers to immunopathology in the absence of infection
- if the disease is driven by components of the innate immune system, it is described as auto-inflammatory
- if it is driven by abnormalities in components of the adaptive immune response, it is described as autoimmune

auto-inflammatory:
- local factors at sites predisposed to disease lead to activation of innate immune cells such as macrophages and neutrophils, which result in tissue damage
- usually localised (e.g., ankylosing spondylitis)

autoimmune disease:
- aberrant T and B cell responses in primary and secondary lymphoid organs leads to breaking of tolerance with the development of immune-reactivity towards self-antigens
- the adaptive immune response plays the predominant role in the clinical expression of disease
- organ-specific antibodies may be present for a long time before the diseases manifest

123
Q

monogenic auto-inflammatory diseases

A
  • mutations in gene encoding a protein involved in a pathway associated with innate immune cell function
  • this often leads to abnormal signalling via key cytokine pathways involving TNF-alpha and/or IL1
124
Q

the inflammasome complex

A
  • the pathway is activated by toxins, pathogens and urate crystals
  • these act via cryopyrin and ASC to activate procaspase 1
  • the activation of procaspase 1 leads to productin of IL1 and NFkappaB (which is a transcription factor that regulates the expression of genes including TNF-alpha)
  • pyrin-marenostrin is a negative regulator of this pathway
    So:
  • a gain of function in cryopyrin will lead to more inflammation
  • a loss of function mutation in Pyrin-Marenostrin will lead to more inflammation
125
Q

Familial Mediterranean Fever

A
  • autosomal recessive
  • mutation in MEFV gene
  • gene encodes Pyrin-Marenostrin
  • Pyrin-Marenostrin is mainly expressed by neutrophils
  • Defective gene leads to failure to regulate cryopyrin driven activation of neutrophils

clinical presentation:
periodic fevers lasting 48-96 hrs associated with:
- abdominal pain due to peritonitis
- chest pain due to pleurisy/pericarditis
- arthritis
- rash
associated with long-term risk of AA amyloidosis:
- the liver produces serum amyloid A as an acute phase protein
- serum amyloid A then deposits in the kidneys, liver and spleen
- deposition in the kidneys is most problematic because it can lead to nephrotic syndrome and renal failure

treatment:
- colchicine
(binds to tubuline in neutrophils and disrupts neutrophil functions including migration and chemokine secretion)

126
Q

monogenic autoimmune diseases

A

caused by mutation in gene encoding a protein involved in a pathway associated with adaptive immune cell function

3 types of pathogenesis:
- abnormality in tolerance
- abnormality in regulatory T cells
- abnormality of lymphocyte apoptosis

127
Q

autoimmune polyendocrinopathy candidiasis ectodermal dystrophy syndrome (APECED)

A
  • autosomal recessive
    defect in autoimmune regulator (AIRE):
  • this is a transcription factor that is vital in the development of T cell tolerance in the thymus
  • it upregulates the expression of self-antigens by thymic cells which T cells are selected against
  • it promotes the apoptosis of auto-reactive T cells

defect in AIRE leads to failure of central tolerance and the release of auto-reactive T cells

this disease can cause multiple autoimmune conditions:
- hypoparathyroidism
- addison’s
- hypothyroidism
- diabetes mellitus
- vitiligo
- enteropathy

these patients are also predisposed to candidiasis

this is because they produce antibodies against cytokines (IL17 and IL22) which increases their risk of candidiasis infection

128
Q

IPEX

A
  • immune dysregulation, polyendocrinopathic, enteropathy, X-linked syndrome
  • caused by mutations in Foxp3 (Forkhead box p3)
  • this is required for the development of Treg cells
  • the lack of Treg cells means that these patients fail to negatively regulate T cell responses, leading to autoantibody formation
  • these patients end up developing autoimmune diseases: enteropathy, diabetes mellitus, hypothyroidism, dermatitis
129
Q

ALPS

A
  • autoimmune lymphoproliferative syndrome
  • due to mutations in the FAS pathway
  • this leads to a defect in apoptosis of lymphocytes
  • in turn, this will cause a failure of tolerance (as autoreactive lymphocytes do not die by apoptosis) and there is a failure of lymphocyte homeostasis

clinical phenotype:
- high lymphocyte count
- large spleen and large lymph nodes
- autoimmune diseases (commonly autoimmune cytopaenias)
- lymphoma

130
Q

polygenic auto-inflammatory disorders

A
  • mutations in genes encoding proteins involved in pathways associated with innate immune cell function
  • they also tend to be slightly localised
  • local factors at predisposed sites can lead to activation of innate immune cells such as macrophages and neutrophils, with resulting tissue damage
  • as these diseases are mainly to do with the innate immune system, their HLA associations are less strong
  • in general, these diseases are NOT characterised by the presence of autoantibodies
131
Q

crohn’s disease - best known loci

A

IBD1 on chromosome 16
NOD2 gene (or CARD-15)

132
Q

crohn’s treatment

A
  • corticosteroids
  • azathioprine
  • anti-TNFalpha antibodies
  • anti-IL12/23 antibodies
133
Q

mixed pattern diseases

A
  • caused by mutations in genes encoding proteins involved in pathways associated with innate AND adaptive immune cell function
  • HLA associations may be present
  • auto-antibodies are NOT usually a feature
134
Q

ankylosing spondylitis

A

highly heritable >90%
around 50% of the heritability of AS comes down to HLA-B27

presentation:
- low back pain and stiffness
- enthesitis
- large joint arthritis

treatment:
- NSAIDs
- immunosuprresion (antiTNF, anti-IL17)

135
Q

role of HLA-B27

A

HLA-B27 presents antigents to CD8 T cells and it is also a ligand for the killer immunoglobulin receptor

  • it tends to occur at specific sites where there are high tensile forces (entheses - sites of insertion of ligaments or tendons)
136
Q

polygenic autoimmune disease

A
  • mutations in genes encoding proteins involved in pathways associated with adaptive immune cell function
  • HLA associations are common
  • aberrant B and T cell responses in primary and secondary lymphoid organs lead to breaking of tolerance with development of immune reactivity towards self-antigens
  • auto-antibodies are found
137
Q

genetic polymorphisms - T cell activation

A
  • PTPN22 suppresses T cell activation
  • so, if it becomes mutated you will fail to control T cell activation leading to the presence of more reactive T cells
  • this will predispose you to the development of autoimmune disease
  • CTLA4 is involved in regulating T cell function
138
Q

Gel and Coombs - effector mechanisms of immunopathology

A
  • Gel and Coombs classified skin test hypersensitivity reactions according to type of immune response observe
  • this classification was broadly looking at whether a response was antibody or T cell mediated
  • these are the effector mechanisms for immunopathology

Gel and Coombs classification:
- type I - immediate hypersensitivity which is IgE mediated
- type II - antibody reacts with cellular antigen
- type III - antibody reacts with soluble antigens to form an immune complex
- type IV - delayed-type hypersensitivity. T cell mediated response

139
Q

IgE mediated reactions (type I)

A
  • rapid allergic reaction
  • pre-existing IgE antibodies to allergen
  • IgE bound to Fc epsilon receptors on mast cells and basophils
  • cell degranulation
  • release of inflammatory mediators
  • increased vascular permeability, leukocyte chemotaxis, smooth muscle contraction
  • this is almost always to FOREIGN antigens
  • however, it is possible that there is some involvement of self-antigens in this type of reaction in some cases of eczema
140
Q

antibody driven immune reactions - hypersensitivity type II

A
  • this is when antibodies bind to cell-associated antigens
  • this is basically always autoimmune
  • binding of antibodies to a cell can lead to an antibody-dependent destruction

antibodies can activate complement (by binding to C1), antibodies can also bind to receptors on NK cells or macrophages which will also result in cell death, binding of antibodies could also lead to receptor activation or blockade (sometimes considered type V responses)

e.g., in Graves’ disease, the antibodies will bind to the TSH receptor and stimulate it
they can also bind to the receptor and block its normal function (e.g., Myasthenia gravis)

141
Q

type III hypersensitivity - immune complex driven

A
  • antibody binds to soluble antigen to form circulating immune complex
  • these antibodies can be generated against endogenous proteins (e.g., anti-nuclear antibodies) or to exogenous substances (e.g., drugs)
  • the immune complexes deposit in the blood vessels (esp. in the kidneys, joints and skin)
  • through their Fc portion, the antibodies can activate complement and activate inflammatory cells
  • this can also lead to tiny bleeds in various places which gives rise to the purpuric rash

examples: SLE, RA

142
Q

type IV hypersensitivity reactions in autoimmunity: T cell driven

A
  • HLA class I molecules present antigens to CD8 T cells
  • CD8 cells will recognise self-peptides presented by HLA Class I molecules leading to damage to cells
  • you can also have CD4 cells recognising peptides presented by HLA Class II molecules
  • this results in cytokine production, which then affects immune function

e.g., insulin dependent diabetes mellitus, RA, multiple sclerosis, experimental autoimmune encephalitis (EAE)

143
Q

recap of immune responses

A
  • pathogens have conserved structures that can be recognised by cells of the immune system (Th1 and Th17)
  • multicellular organisms and allergens don’t necessarily have conserved structures that are recognised by immune cells, instead they release mediators (e.g., proteases) that disturb epithelial barriers which is a functional change that is recognised by the immune system and gives rise to Th2-mediated responses
144
Q

overview of the Th2 immune response

A
  • stressed/damaged epithelium will release signalling cytokines (e.g., TSLP)
  • these cytokines will act on Th2 cells, Th9 cells and ILC2 cells and promote the section of IL4, IL5 and IL13
  • these then act on eosinophils and basophils which plays a role in the expulsion of parasites and allergens but can also contribute to tissue injury
  • the TSLP and other cytokines released by the damaged epithelium can also activate follicular Th2 cells which then releases IL4
  • IL4 stimulates B cells to produce IgE and IgG4
145
Q

induction of Th2 immune responses

A
  • not well understood
  • the primary defect is through the epithelial barrier e.g., skin defect is a significant risk factor for the development of IgE antibodies (atopic dermatitis)
  • skin dendritic cells (Langerhans cells and dermal dendritic cells) promote secretion of Th2 cytokines much more efficiently than other dendritic subtypes
  • this suggests that different dendritic cell subsets will prime the Th2 immune responses in humans to different levels
  • IL4 secretion is only induced by peptide-MHC presentation to TCR or naive/memory Th2 cells

take home message: oral exposure promotes immune tolerance whereas skin and respiratory exposure induces IgE sensitisation

146
Q

age of onset of allergic diseases

A

infants
- atopic dermatitis
- food allergy (milk, egg, nuts)

childhood
- asthma (house dustmite, pets)
- allergic rhinitis

adults
- drug allergy
- bee allergy
- oral allergy syndrome
- occupational allergy

147
Q

investigation of allergic diseases

A

elective investigations
- skin prick and intradermal tests
- laboratory measurement of allergen-specific IgE
- component-resolved diagnostics
- basophil activation test
- challenge test (supervised exposure to antigen)

during acute episode:
evidence of mast cell degranulation:
- serial mast cell tryptase
- blood and/or urine histamine

148
Q

specific IgE sensitisation tests

A
  • skin prick and blood tests are used to detect the presence/absence of IgE antibody against external proteins
  • a positive IgE test only demonstrates sensitisation not clinical allergy

risk profile of serum IgE for prediction of allergic symptoms:
- concentration - higher levels = more symptoms
- affinity to target - higher affinity = increased risk
- capacity of IgE antibody to induce mast cell degranulation

detection of IgE is necessary but not sufficient to make a diagnosis of allergic disease
- diagnosis requires history, examination, blood tests, skin prick tests etc to be combined

149
Q

advantages & disadvantages of skin prick test

A

advantagesL
- rapid (read after 15-20 mins)
- cheap and easy to do
- excellent negative predictive value (>95%)
- increasing size of wheals correlates with higher probability for allergy
- patient can see the response

disadvantages:
- requires experience to interpret
- risk of anaphylaxis (1 in 3000)
- poor positive predictive value: high false positive rate
- limited value in patients with dermatographism or extensive eczema
- false negative results with labile commercial food extracts

150
Q

indications for serum specific IgE blood test

A
  • patients who can’t stop antihistamines
  • patients with dermatographism
  • patients with extensive eczema
  • history of anaphylaxis
  • borderline/equivocal skin prick test results
151
Q

component resolves diagnostics (CRD) (+ indications for allergen component testing)

A
  • a blood test to detect IgE to single protein components - abundance and stability of protein contributes to risk of allergic disease
  • useful for peanut and hazelnut allergy (may reduce need for food challenges)

indications:
- detect primary sensitisation
- confirm cross-reactivity
- define risk of serious reaction for stable allergens
- improve diagnostic sensitivity on addition of components which are poorly represented in whole food extracts
- improve diagnostic sensitivity for unstable molecules in whole food extracts

152
Q

mast cell tryptase: a biomarker for anaphylaxis

A
  • tryptase is a pre-formed protein found in mast cell granules
  • systemic degranulation of mast cells during anaphylaxis results in increased serum tryptase
  • peak concentration = 1-2 hours
  • baseline = 6-12 hours
  • failure of return to baseline after anaphylaxis may be indicative of systemic mastocytosis
  • useful if the diagnosis of anaphylaxis is uncertain (e.g., hypotension and rash during anaesthesia)
  • reduced sensitivity for food-induced anaphylaxis
153
Q

challenge tests: food and drug allergy

A
  • gold standard for food and drug allergy diagnosis
  • increasing volumes of the offending food/drug are ingested
  • double-blind placebo or open challenge
  • take place under close medical supervision
  • difficult to interpret mild symptoms
  • risk of severe reaction
154
Q

basophil activation test

A
  • measurement of basophil response to allergen IgE cross-linking
  • activated basophils increase the expression of CD63, CD203 and CD300 protein on their cell surface
  • increasingly used in the diagnosis of food and drug allergy
155
Q

IgE anaphylaxis - cells, mediators, examples

A

cells:
- mast cells
- basophils

mediators:
- histamine & PAF

examples:
- food, insect venom, ticks, penicillin

156
Q

IgG anaphylaxis - cells, mediators, examples

A

cells:
- macrophages
- neutrophils

mediators:
- histamine & PAF

examples:
- biologicals, blood and IgG transfusions

157
Q

complement anaphylaxis - cells, mediators, examples

A

cells:
- mast cells
- macrophages

mediators:
- PAF & histamine

examples:
- lipid excipients, liposomes, dialysis membranes and PEG

158
Q

pharmacological anaphylaxis - cells, mediators, examples

A

cells:
- mast cells

mediators:
- leukotrienes & histamine

examples
- NSAID inc. aspirin, opiates, neuromuscular and quinolones drug

159
Q

anaphylaxis - mechanism of adrenaline

A

alpha 1 - causes peripheral vasoconstriction, reverses low BP and mucosal oedema

beta 1 - increases heart rate, contractility and BP

beta 2 - relaxes bronchial smooth muscle and reduces the release of inflammatory mediators

160
Q

management of anaphylaxis

A

IM adrenaline!

supportive:
- adjust body position
- 100% oxygen
- fluid replacement
- inhaled bronchodilators
- hydrocortisone 100mg IV (prevent late phase response)
- chlorpheniramine 10mg IV

further:
- referral to allergy/immunology clinic
- investigate cause
- written info on recognition of symptoms, avoidance of triggers, indications for self-treatment with EpiPen
- prescription of emergency kit to manage anaphylaxis
- review patient’s understanding

161
Q

immune response to transplanted graft

A

phase 1: recognition of foreign antigens
phase 2: activation of antigen-specific lymphocytes
phase 3: effector phase of graft rejection

most relevant protein variations in clinical transplantation are:
- ABO blood group
- HLA (coded on chromosome 6 by MHC)
[there are some other minor histocompatibility genes]

2 major components to rejection:
1) T-cell mediated rejection
2) antibody-mediated rejection

162
Q

HLA - classes and variability

A
  • HLA Class I (A, B and C); expressed on ALL cells
  • HLA Class II (DR, DQ, DP); expressed on antigen-presenting cells but can also be upregulated on other cells under stress
  • they are highly polymorphic with hundreds of alleles for each locus
  • the areas of protein lining the peptide-binding groove are responsible for the high degree of variability between HLA
  • the high variability has evolved so that we are able to present a wide variety of antigens in that peptide-binding groove to the cells of the immune system
  • the variability in antigens is an issue in transplantation because they provide a key difference that the immune system can react with
  • A, B and DR are thought to be the most immunogenic
  • the number of mismatches is a major determinant of the risk of rejection
163
Q

T cell mediated transplant rejection - effector phase

A
  • the T cells will tether, roll and arrest on the endothelial cell surface
  • they will then crawl through into the interstitium and start attacking the tubular epithelium
  • typical histological features of T cell-mediated rejection:
    a) lymphocytic interstitial infiltration
    b) ruptured tubular basement membrane
    c) tubulitis (inflammatory cells within the tubular epithelium)
  • macrophages, recruited by the T cells, may also be seen in the interstitium and the tubules
164
Q

antibody-mediated transplant rejection

A

phase 1: exposure to foreign antigen
phase 2: proliferation and maturation of B cells with antibody production
phase 3: effector phase - antibodies bind to graft endothelium (capillaries of glomerulus and around tubules)

  • we have naturally occurring anti-A and anti-B antibodies
  • whereas anti-HLA antibodies are NOT naturally occurring:
    a) they can be pre-formed due to previous exposure to epitopes (e.g., previous transplantation, pregnancy, transfusion)
    b) or they can be post-formed - formed after transplantation
165
Q

role of antibodies in rejection of transplant

A
  • antibodies will bind to antigens (HLA) on the endothelium of the blood vessels in the transplanted organ
  • these antibodies can then fix complement which assembles to form membrane attack complexes (MAC) resulting in endothelial cell lysis
  • binding of complement can also recruit inflammatory cells to the microcirculation
  • the antibodies can also directly recruit mononuclear cells, NK cells and neutrophils
  • one of the cardinal features of antibody-mediated rejection is the presence of inflammatory cells within the capillaries of the kidney which then cause endothelial injury (capillaritis)
  • these processes will result in procoagulant tendencies and closure of the microcirculation leading to graft fibrosis
  • antibodies against graft endothelial epitopes can also cause damage by cross-linking the MHC molecules and activating them
166
Q

prevention and treatment of graft rejection

A

1) AB/HLA typing
- part of the allocation procedure
- encourage living donation from blood relatives
- HLA matching is particularly important for bone marrow and kidney transplantation
- it is less important in heart and lung transplants
- DNA sequencing using PCR is how people are typed

2) screening for antibodies
- before transplantation
- at the time of transplantation (once organ has been assigned)
- after transplantation (repeat measurement to check for new antibody formation)

3) overcoming organ mismatch issues
- improve transplantation across tissue barriers
- more donors
- organ exchange programmes
- future: xenotransplantation (animals), stem cell research

167
Q

3 types of assay for anti-HLA antibodies

A

1) cytotoxicity assays
- looks at whether the recipient’s serum will kill the lymphocytes of the donor in the presence of complement
- positive crossmatch suggests that there is cell lysis

2) flow cytometry
- looks at whether the recipient’s serum binds to the donor’s lymphocytes
- bound antibody is detected by fluorescently-labelled anti-human immunoglobulin
- this is broad and looks at whether the antibodies will bind antigen irrespective of whether they bind complement

3) solid phase assays
- this uses a series of beads containing all the possible HLA epitopes
- the recipient’s serum is mixed with these beads and fluorescently-labelled immunoglobulin is used to determine which HLA epitopes the antibodies bind to
- it can also give an indication of the strength of the reaction
- NB: patients that have antibodies to lots of different types of antibodies are regarded as highly sensitised

168
Q

drugs used for immunosuppression post-transplant that target T cells

A

1) steroids
normally given as part of a standard immunosuppression regime to prevent T-cell mediated rejection

2) calcineurin inhibitors (tacrolimus, cyclosporine)

3) cell cycle inhibitors (mycofenolate mofetil, azathioprine)

4) targeting TCR, causing T cell apoptosis (anti-CD3 antibody, anti-thymocyte globulin)

5) anti-CD52 monoclonal antibodies
- alemtuzumab; causes lysis of T cells
- daclizumab; targets the cytokine signal

169
Q

targeting antibody-mediated rejection in immunosuppression post-transplant

A

main targets:
- B cell activation
- secretion of antibodies by plasma cells
- effect of antibodies on endothelium

  • B cells can be depleted using rituximab (anti-CD20)
  • BAFF inhibitors target cytokines that promote B cell activation and growth
  • proteasome inhibitors such as bortezemib can block the production of antibodies by plasma cells
  • there are also a number of drugs that target the interaction between T cells and B cells
  • complement binding to the surface of endothelial cells can be blocked using complement inhibitors such as eculizimab
170
Q

baseline immunosuppression post-transplant

A

calcineurin inhibitor + mycofenolate mofetil or azathioprine with or without steroids

171
Q

treatment of episodes of acute rejection

A

cellular: steroids, OKT3/ATG

antibody-mediated: IVIG, plasma exchange, anti-C5 and anti-CD20
(NB: IVIG can reduce antibody production through feedback and it can displace troublesome antibodies so that they cannot exert their harmful effects)

172
Q

in which cases is haematopoietic stem cell transplantation used?

A
  • used for haematological and lymphoid cancers
  • may be used in acquired or inherited deficiencies in marrow cells such as errors of metabolism or immunodeficiencies
173
Q

graft-versus-host disease in stem cell transplantation

A
  • during the process of SCT, the host immune system is eliminated (using total body irradiation and drugs)
  • it is then replaced by own (autologous) or HLA-matched donor (allogeneic) bone marrow
  • allogeneic stem cell transplantation leads to reaction of donor lymphocytes against host tissues
  • related to a degree of HLA-incompatibility
  • if there is a malignancy (e.g., leukaemia), the graft can help kill these cells (graft-versus-tumour)

GVHD prophylaxis: methotrexate/cyclosporine

symptoms:
- rash
- N&V
- abdo pain
- diarrhoea/bloody stool
- jaundice

treatment: steroids

174
Q

post-transplantation malignancy

A
  • viral associated malignancies are much more common [Kaposi sarcoma (HHV8), lymphoproliferative disease (EBV)]
  • skin cancer is 20x more common
  • risk of other cancers is also increased
175
Q

HIV target

A

CD4+ T helper cells

176
Q

HIV receptor and co-receptor

A
  • the CD4 molecule is the receptor for HIV-1
  • most infecting strains of HIV-1 will also require co-receptor molecules (CCR5 or CXCR4) to enter the cells
  • there is evidence to suggest that there are more receptors involved in HIV entry (researchers have shown that blocking 3 chemokine receptors could block infection)
177
Q

modes of HIV transmission

A

1) sexual
- the virus enters through mucosal surfaces
- transmission is increased by activities that damage these surfaces (e.g., anal sex)
- CD4+ cells and dendritic cells in the mucosa may bind to the virus and carry it from the site of the infection to the lymph nodes where other immune cells will become infected

2) infected blood
- transfusion
- sharing needles
- blood products

3) mother to child
- before/during birth
- breastfeeding

178
Q

summary of effects of HIV-1 on CD8+ T cells

A
  • CD4+ T cells are disabled (anergised) by the virus
  • therefore, monocytes and dendritic cells are not activated by the CD4+ T cells and cannot prime the naive CD8+ T cells
  • CD8+ T cell and B cell responses are diminished without help from CD4+ T helper cells
  • CD4+ T cell memory is lost
  • infected monocytes and dendritic cells will be killed by the virus or by CD8+ T cells
  • they fail to activate the memory of CTL
179
Q

HIV - variation and mutation of the viral genome

A

replication of the HIV genome is dependent on 2 steps during which errors can occur:
- reverse transcriptase (RNA to DNA) - lacks proof-reading mechanisms associated with cellular DNA polymerases and therefore genomes of retroviruses are copied to DNA with low fidelity
- transcription of DNA into RNA copies - also low fidelity

this means that HIV can accumulate a lot of mutations with numerous variants or quasispecies
the propensity to mutate can lead to the development of advantageous features such as:
- escape from neutralising antibodies
- escape from HIV-1 specific T cells
- resistance and escape from antiretroviral drugs

180
Q

life cycle of HIV

A

1) attachment and entry
2) reverse transcription and DNA synthesis
3) integration
4) viral transcription
5) viral protein synthesis
6) assembly of virus and release of virus
7) maturation

181
Q

points/processes in HIV life cycle that can be targeted therapeutically

A
  • attachment
  • fusion
  • reverse transcription
  • integration of viral DNA into host DNA
  • transcription of DNA to viral RNA
  • translation of viral RNA to produce viral proteins
  • viral protein cleavage by proteases
  • assembly and budding of new HIV
182
Q

clinical course of HIV

A
  • median time from HIV infection to development of AIDS = 8-10 years
  • viral burden predicts disease progression
  • rapid progressors (10%) will take 2-3 years
  • long-term non-progressors (LTNP < 5%) will have stable CD4+ counts and no symptoms after 10 years
  • HAART significantly improves prognosis
  • exposed-seronegatives (ESN) are people who are repeatedly exposed to HIV but do not seroconvert
183
Q

possible mechanisms of HIV long-term non-progression

A

host genetic factors:
- slow progressor HLA profile
- heterozygosity for 32-bp deletion in chemokine receptor CCR5
- mannose binding lectin alleles
- tumour necrosis factor c2 microsatellite alleles
- Gc vitamin D-binding factor alleles

host immune response factors:
- effective CTL & HTL responses
- secretion of CD8 antiviral factor
- secretion of chemokines that block HIV entry co-receptors CCR5 and CXCR4
- secretion of IL-16
- effective humoral immune response
- maintenance of functional lymphoid tissue architecture

virologic factors:
- infection with attenuated strains of HIV

184
Q

detection of HIV-1

A
  • detect the presence of anti-HIV antibodies (ELISA)
  • detect viral load (viral RNA using PCR)
  • HIV anitbody ELISA = screening test
  • HIV antibody Western blot = confirmatory test
  • initial baseline plasma viral load is a good predictor of time taken for the disease to appear
  • CD4+ T cell counts (using flow cytometry)
185
Q

HIV resistance testing

A

phenotypic
- viral replication is measured in cell cultures under selective pressure of increasing concentrations of antiretroviral drugs (compared to wild-type)

genotypic
- mutations detected by direct sequencing of the amplified HIV genome (so far limited to sequencing RT and P)

186
Q

aim of highly active antiretroviral therapy (HAART)

A
  • substantial control of viral replication
  • increase in CD4+ T cell counts
    a. initial rise due to redistribution of memory T cells
    b. later rise is due to thymic naive T cells
  • improvement in their host defences (decline in opportunistic infections)

NB: HAART does not eliminate HIV from the body because there is a reservoir in CD4+ T cells

187
Q

when to start HAART?

A
  • all symptomatic patients
  • all CD4+ count < 200 cells/microL
  • CD4 count 200-350 cells/microL

general formula:
- 2 x NRTI (nucleoside reverse transcriptase inhibitors)
- 1 X NNRTI (NON-NRTI) or boosted PI (protease inhibitor)

188
Q

limitations and complications of HAART

A
  • does NOT eradicate latent HIV-1
  • fails to restore HIV-specific T cell responses
  • threat of drug resistance
  • significant toxicities
  • high pill burden
  • adherence
  • quality of life
  • cost
189
Q

mantoux test

A
  • inject a small amount of liquid tuberculin (aka purified protein derivativ (PPD)) intradermally
  • the area of injection is examined 48-72 hours after tuberculin injection
  • the reaction is an area of swelling around the injection site
  • protection usually lasts for 10-15 years
190
Q

examples of live attenuated vaccines

A

MMR, BCG, yellow fever, typhoid, polio (sabin), vaccinia

191
Q

advantages and disadvantages of live attenuated vaccines

A

advantages:
- establishes infections (ideally mild)
- raises broad immune response to multiple antigens (more likely to offer protection against different strains)
- activates all phases of immune system
- often confer life-long immunity after 1 dose

disadvnatages:
- storage problems
- possible reversion to virulence
- spread to contacts
- spread to immunocompromised/immunosuppressed

192
Q

examples of inactivated/component vaccines

A

influenza, cholera, polio (Salk), Hep A, pertussis, rabies
toxoids (inactivated toxins): diptheria, tetanus

193
Q

advantages and disadvantages of inactivated/component vaccines

A

advantages:
- no mutation or reversion
- can be used in immunodeficient patients
- can lead to elimination of wild-type virus from the community
- easier storage
- lower cost

disadvantages:
- often do not follow normal route of infection
- may have poor immunogenicity
- may need multiple injections
- may require conjugates or adjuvants

194
Q

what is a conjugate vaccine? mechanism? examples?

A
  • polysaccharide + protein carrier
  • the polysaccharide alone induces a T cell-independent B cell response (transient)
  • addition of the protein carrier promotes T cell immunity which enhances the B cell/antibody response

examples:
- haemophilus influenzae B
- meningococcus
- pneumococcus
NB: these are polysaccharide encapsulated bacteria

195
Q

role of adjuvants in vaccines + examples

A
  • increase the immune response without altering its specificity
  • these mimic the action of PAMPs on TLR and other PRRs

examples:
- aluminium salts
- lipids - monophosphoryl lipid A
- Freund’s adjuvant (in animals)

196
Q

dendritic cell vaccines

A
  • this is used against tumours where dendritic cell function may be compromised
  • you take a patient’s dendritic cells and load them with a tumour antigen and reintroduce them to the patient to try and boost the immune response against hte tumour antigens
  • this requires antigens that are specific to the tumour and distinct from normal cells
197
Q

indications for antibody replacement

A

primary antibody deficiency
- X-linked agammaglobulinaemia
- X-linked hyper-IgM syndrome
- common variable immunodeficiency

secondary antibody deficiency:
- haematological malignancies (CLL, multiple myeloma)
- after BM transplant

198
Q

cellular immunotherapy for EBV-associated disease

A
  • can be used for EBV in people who are immunosuppressed to prevent the development of B cell lymphoproliferative disease
  • they need function EBV-specific T cells
  • blood is taken from the patient or from a matched individual
  • the peripheral blood mononuclear cells are isolated
  • they are then stimulated with EBV peptides
  • this creates an expansion of EBV-specific T cells
  • they are then infused back to the patient
199
Q

tumour infiltration T cell therapy

A
  • you remove the tumour from the patient
  • you stimulate the T cells within the tumour with cytokines (e.g., IL-2) in the presence of the tumour so that they develop a response against the tumour
  • you then select and expand the tumour infiltrating lymphocytes and reinfuse into the patient
200
Q

TCR and CAR T cell therapy

A
  • T cells are taken from the patient and viral or non-viral vectors are used to insert fragments of genes into these T cells
  • these gene fragments encode receptors
  • for TCR therapy, you will insert a gene that encodes a specific TCR (e.g., against a tumour cell antigen)
  • in CAR therapy, the receptors are chimeric (it contains both B ant T cell components)
  • the type of CAR therapy that has come into use is targeting CD19 (which is present on B cells)
  • the receptors on the CAR cell has an immunoglobulin variable domain at the end and it is joined onto the remainder of the T cell receptor (CD28 + CD3)
  • so, it signals through the usual TCR pathway but it recognises CD19 through an immunoglobulin domain
  • this recognises CD19 on B cells and harnesses the T cells to kill the B cells
  • this is increasingly being used in ALL and NHL
201
Q

biologics used to block immune checkpoints

A

ipilimumab - antibody specific to CTLA4

pembrolizumab and nivolumab - antibodies specific or PD-1

202
Q

clinical use of recombinant cytokines

A

the aim is to modify the immune response

interferon alpha
- used as an adjunct in the treatment of Hep B, Hep C, Kaposi sarcoma, CML, multiple myeloma

interferon beta
- Behcet’s disease
- relapsing multiple sclerosis (no longer in use)

interferon gamma
- chronic granulomatous disease

203
Q

approaches to suppressing the immune system

A
  • steroids
  • anti-proliferative agents
  • plasmapheresis
  • inhibitors of cell signalling
  • agents directed against cell surface antigens
  • agents directed at cytokines
204
Q

effects of corticosteroids on prostaglandins

A
  • corticosteroids inhibit phospholipase A2
  • phospholipase A2 is usually involved in the conversion of phospholipids into arachidonic acid which is then converted to eicosanoids (e.g., prostaglandins and leukotrienes) by COX
  • by inhibiting phospholipase A2, corticosteroids will block arachidonic acid and prostaglandin formation thereby reducing inflammation
205
Q

effects of corticosteroids on phagocytes

A

decrease traffic of phagocytes to inflamed tissue
- reduce the expression of adhesion molecules on the endothelium
- they also block the signals that tell immune cells to move from the bloodstream into tissues
- this leads to a transient increase in neutrophil count

decrease phagocytosis
decrease release of proteolytic enzymes

206
Q

effects of corticosteroids on lymphocyte function

A
  • lymphopaenia (sequestration of lymphocytes into lymphoid tissue: affects CD4>CD8>B cells)
  • blocks cytokine gene expression
  • decreased antibody production
  • promotes apoptosis
207
Q

what are anti-proliferative agents? examples? mechanism of action?

A

agents that inhibit lymphocyte proliferation
- cyclophosphamide
- mycophenolate
- azathioprine
- methotrexate

action:
- inhibit DNA synthesis
- cells with rapid turnover are most sensitive

208
Q

cyclophosphamide - mechanism, indications, side effects

A

mechanism:
- alkylates guanine base of DNA
- damages DNA and prevent cell replication
- affects B cells > T cells (tends to be used in antibody-mediated disorders)
- at high doses, affects all cells with high turnover

indications:
- multisystem connective tissue disease
- vasculitis with severe end-organ involvement (e.g., GPA, SLE)
- anti-cancer agent

side effects:
- BM suppression
- sterility (mainly males)
- hair loss
- haemorrhagic cystitis
- bladder cancer
- haematological malignancies
- non-melanoma skin cancer
- teratogenic
- infection (e.g., pneumocystic jirovecii)

209
Q

azathioprine - mechanism, indications, side effects

A

mechanism:
- metabolised by the liver to 6-mercaptopurine
- blocks de novo purine synthesis (e.g., adenine and guanine)
- prevents replication of DNA
- preferentially inhibits T cell activation and proliferation

indications:
- transplantation
- auto-immune disease
- auto-inflammatory disease

side effects:
- BM suppression
- hepatotoxicity
- infection

210
Q

mycophenolate mofetil - mechanism, indications, side effects

A

mechanism:
- blocks de novo nucleotide synthesis
- prevents replication of DNA
- prevent T cell > B cell proliferation

indications:
- widely used in transplantation (alternative to azathioprine)
- also used in autoimmune disease and vasculitis as an alternative to cyclophosphamide

side effects:
- bone marrow suppression
- teratogenic
- infection

211
Q

inhibitors of cell signalling

A
  • calcineurin inhibitors
  • JAK inhibitors
  • PDE4 inhibitors
212
Q

calcineurin inhibitors

A
  • inhibitors of calcineurin (e.g., ciclosporinand tacrolimus) will prevent T cell signalling
  • therefore, it blocks IL2 production
  • IL2 normally acts on T cells and drives proliferation
  • so, calcineurin inhibitors will prevent T cell activation and proliferation

examples: ciclosporin, tacrolimus

side effects: nephrotoxicity, hypertension, neurotoxic, diabetogenic (tacrolimus+), dysmorphic features (ciclosporin only)

NB: monitor BP and renal function

213
Q

JAK inhibitors

A
  • example: tofacitinib (JAK1 and JAK3 inhibitor)
  • interferes with JAK-STAT signalling (important in transducing the signals from cytokine binding)
  • influences gene transcription
  • inhibits the production of inflammatory molecules
  • effectives in rheumatoid arthritis
214
Q

PDE4 inhibitors

A
  • example: apremilast
  • phosphodiesterase 4 (PDE4) is important in the metabolism of cAMP
  • leads to increase in cAMP
  • cAMP activates PKA which prevent activation of transcription factors
  • this leads to a decrease in cytokine production
  • effective in psoriasis and psoriatic arthritis
215
Q

(rabbit) anti-thymocyte globulin

A
  • thymocyte (lymphocytes from the thymus gland) from humans were injected into rabbits
  • the rabbits then produced antibodies against the thymocytes of varying specificities (e.g., antibodies to CD2, CD3, CD4, CD8 etc)
  • the serum was then taken and injected into patients
  • this is very effective at targeting T cells, however, it is very non-specific
  • the main aim is to cause T cell depletion (thereby reducing the activation and migration of T cells)
  • this is useful in allograft rejection

side effects:
- infusion reactions
- leukopaenia
- infection
- malignancy

216
Q

basiliximab - anti-CD25

A
  • used for prophylaxis of allograft rejection
  • used before and after transplant surgery
  • basiliximab targets part of the IL2 receptor
  • this leads to inhibition of T cell proliferation

side effects:
- infusion reaction
- infection
- concern of long term malignancy risk

217
Q

abatacept - CTLA4-Ig fusion protein

A
  • abatacept is a receptor that is made from a fusion of CTLA4 and IgG Fc component
  • APCs have CD80 and CD86 which can bind to receptors on T cells
    CD28 - activating singal
    CTLA4 - inhibitory signal
  • abatacept will bind to CD80 and CD86, thereby meaning that these receptors can no longer engage with CD28 and CTLA4 to activate/regulate the T cell response
  • this results in reduced T cell activation
  • it is effective in rheumatoid arthritis

side effects:
- infusion reactions
- infection (TB, HBV, HCV)
- caution with malignancy

218
Q

rituximab - anti-CD20 antibodies

A
  • CD20 is expressed on mature B cells but NOT plasma cells
  • this leads to depletion of mature B cells

indications:
- lymphoma
- rheumatoid arthritis
- SLE

  • given as 2 IV doses every 6-12 months (in RA)

side effects:
- infusion reactions
- infection (PML)
- exacerbation of cardiovascular disease

219
Q

agents directed at cell surface antigens + action

A

drugs:
- rabbit anti-thymocyte globulin
- basiliximab - anti-CD25
- abatacept - CTLA4-Ig
- rituximab - anti-CD20
- natalizumab - anti-alpha4 integrin
- tocilizumab - anti-IL-6 receptor

action:
- block signalling
- cell depletion

220
Q

natalizumab - anti-alpha4 integrin

A
  • alpha 4 is expressed with either beta-1 or beta-7 integrin
  • this complex binds to VACM1 or MadCAM1 to mediate rolling and arrest of leukocytes
  • the complex can also bind to non-endothelial VCAM1 in lymphoid tissue
  • therefore, blocking this integrin can inhibit leukocyte migration
  • used in highly-active relapsing-remitting multiple sclerosis

side effects:
- infusion reactions
- infection (PML)
- hepatotoxic
- concerns regarding malignancy

221
Q

tocilizumab - anti-IL6 receptor

A

leads to reduced activation of macrophages, T cells, B cells and neutrophils

indications:
- Castleman’s disease (IL6 producing tumour)
- rheumatoid arthritis

side effects:
- infusion reactions
- injection
- hepatotoxicity
- elevated lipids
- caution with malignancy

222
Q

agents directed at cytokines

A

drugs:
- infliximab (anti-TNFalpha)
- adalimumab (anti-TNFalpha)
- certolizumab (anti-TNFalpha)
- golimumab (anti-TNFalpha)
- etanercept (TNF receptor p75-IgG fusion protein)
- ustekinumab (anti-IL-12 and IL-23)
- secukinumab (anti-IL-17)
- denosumab (anti-RANK ligand)

action:
- block action of cytokines

223
Q

anti-TNFalpha antibodies - infliximab, adalimumab, certolizumab, golimumab

A
  • inhibit TNFalpha
  • TNF-alpha is a critical molecule within the cytokine cascade that is responsible for the inflammatory response in inflammatory arthritis

indications:
- rheumatoid arthritis
- ankylosing spondylitis
- psoriasis and psoriatic arthritis
- inflammatory bowel disease

side effects:
- infusion or injection site reactions
- infection (TB, HBV, HCV)
- lupus-like conditions
- demyelination
- malignancy

224
Q

etanercept - TNF-alpha antagonist

A
  • it is essentially a decoy receptor that mops up TNF-alpha
  • this inhibits the action of TNF-alpha and TNF-beta
  • it is given as a subcutaneous injection

indications:
- rheumatoid arthritis
- ankylosing spondylitis
- psoriasis and psoriatic arthritis

side effects:
- injection site reactions
- infection (TB, HBV, HCV)
- lupus-like conditions
- demyelination
- pregnancy

225
Q

ustekinumab - antibody to p40 subunit of IL12 and IL23

A

the p40 subunit is present in both IL12 and IL23

indications:
- psoriasis and psoriatic arthritis
- crohn’s

  • this leads to inhibition of IL12 and IL23
  • these cytokines mainly act on T cell and NK cells thereby modulating their activity

side effects:
- injection site reactions
- infection (TB)
- concern about malignancy

226
Q

secukinumab - antibody to IL17A

A

dimer of IL17A or IL17A/F will bind to IL17RA/RC receptor

indications:
- psoriasis and psoriatic arthritis
- ankylosing spondylitis

leads to inhibition of IL17A

side effects:
- infection (TB)

227
Q

denosumab - antibody against RANKL

A
  • RANKL is produced by osteoblasts and it acts on RANK (receptor) on osteoclasts
  • the binding of RANKL to RANK leads to osteoclast differentiation and function
  • this, therefore, leads to increased bone resorption
  • there is a natural decoy receptor called osteoprotergin (OPG) which can bind to RANKL and regulate the system
  • denosumab is an antibody directed against RANKL which prevents RANKL from binding to RANK on osteoclasts, thereby reducing osteoclast differentiation and function (and, therefore, reducing bone resorption)

indications:
- osteoporosis
- given subcutaneously every 6 months

side effects:
- injection site reactions
- infection
- avascular necrosis of the jaw

228
Q

side effects of biologic agents

A

infusion reactions:
- urticaria, hypotension, tachycardia, wheeze - IgE mediated
- headaches, fever, myalgias - not classical type I hypersensitivity
- cytokine storm

injection site reactions:
- peak reaction at 48 hrs
- may also occur at previous injection sites (recall reactions)
- mixed cellular infiltrates, often with CD8 T cells
- not generally IgE or immune complexes

acute infection:
- risk often >2x backrground
- avoidance
- vaccination (NB: do not give live vaccines to immunosuppressed patients)
- temporarily stop immunosuppression
- consider atypical organisms
- appropriate antibiotics

229
Q

Graves’ disease

A
  • excessive production of thryoid hormones
  • mediated by IgG antibodies that stimulate the TSH receptor
  • the antibodies bind to the TSH receptor and stimulate it leading to overproduction of thyroid hormones
  • negative feedback does NOT override antibody stimulation
  • this is a type II hypersensitivity reaction (however, it is stimulatory rather than destructive)
230
Q

Hashimoto’s thyroiditis

A
  • most common cause of hypothyroidism is iodine-replete areas
  • may present with a goitre - enlarged thyroid infiltrated by T and B cells
  • associated with anti-TPO antibodies (thyroid damage & lymphocyte inflammation)
  • associated w the presence of anti-thyroglobulin antibodies
  • type II and type IV hypersensitivity reaction
231
Q

immunology of type 1 diabetes

A

CD8+ T cell infiltration of the pancreas
- the T cell clones have specificity for islet antigens
- the CD8+ lymphocytes bind to peptides presented by MHC class I molecules on the beta-cells of the pancreas
- these autoantigens include GAD and IA2
- the presence of antibodies against these antigens will pre-date the development of the disease:
anti-islet cell
anti-insulin
anti-GAD
anti-IA2
NB: detection of antibodies does NOT currently play a role in diagnosis

232
Q

pernicious anaemia

A
  • antibodies against intrinsic factor which leads to failure of absorption of vitamin B12
  • vitamin B12 deficiency can also lead to subacute degeneration of the spinal cord (this involves the posterior and lateral columns)
  • other neurological features include peripheral neuropathy and optic neuropathy
  • antibodies against gastric parietal cells or intrinsic factor are useful in diagnosis
233
Q

myasthenia gravis

A
  • antibodies against the nicotinic acetylcholine receptor
  • this leads to a failure of depolarisation
  • characterised by fluctuating weakness
  • ptosis is a common feature
  • EMG studies are usually abnormal
  • tensilon test to confirm diagnosis [involves administering a very short-acting anticholinesterase (edrophonium bromide), which will cause a rapid improvement in symptoms]
  • anti-acetylcholine receptor antibodies are present in 75% of patients and so they are useful in diagnosis
  • offspring of affected mothers may experience transient neonatal myasthenia
  • type II hypersensitivity reaction
234
Q

Goodpasture’s syndrome

A
  • aka anti-glomerular basement membrane disease
  • leads to lung and kidney damage
  • the deposition of antibodies along the basement membrane gives a smooth linear appearance
  • they are detected using fluorescein conjugated anti-human immunoglobulin
235
Q

environmental factors associated with rheumatoid arthritis

A
  • smoking is associated with the development of erosive disease (due to increased citrullination)
  • gum infections with Porphyromonas gingivalis is associated with RA
236
Q

antibodies specific to rheumatoid arthritis

A
  • anti-cyclin citrullinated peptide antibodies (95% specific)
  • rheumatoid factor
237
Q

B cell involvement in rheumatoid arthritis pathology

A

type II
- antibodies bind to citrullinated peptides leading to activation of complement, macrophages and NK cells

type III
- immune complexes form and get deposited
- this leads to complement activation

238
Q

T cell involvement in rheumatoid arthritis pathology

A
  • antigen presenting cells will present peptides to CD4 cells which will then lead to the production of IFN-gamma and IL17
  • these cytokines act on fibroblasts and macrophages
  • this, in turn, leads to the production of:
    MMPs
    IL-1
    TNF-alpha
239
Q

the joint in RA

A
  • the synovium becomes very inflamed forming a pannus
  • this invades the articular cartilage and adjacent bone
  • there is also an increase in synovial fluid volume
240
Q

SLE - pathology

A
  • abnormalities in clearing apoptotic cells
  • abnormalities in cellular activation
  • B cell hyperactivity and loss of tolerance
  • antibodies directed particularly at intracellular proteins
  • antibodies bind to antigens forming immune complexes
  • these can deposit in tissues (e.g., skin, joint, kidneys)
  • immune complexes can activate complement via the classical pathway
  • immune complexes can stimulate cells that express Fc and complement receptors
  • immune complex (type III) vs antibody (type II)-mediated disease pathology
241
Q

immunological investigations in SLE

A

anti-nuclear antibody

> 1:640 (normal <1:80)

NB: this is the minimum dilution at which the antibody can be detected

242
Q

targets of anti-nuclear antibodies

A

dsDNA

extractable nuclear antigens (ENAs)
- ribonucleoproteins
- enzymes (e.g., RNA polymerase or topoisomerase)

243
Q

homogenous staining in SLE

A

Homogenous pattern:
- homogenous staining associated with staining for dsDNA
- specificity is confirmed using ELISA
- anti-dsDNA antibodies are highly specific for SLE
- these will be present in 60-70% of SLE patients at some point
- high titres are associated with severe disease, including renal or CNS involvement
- useful in disease monitoring - an increase in antibody titres is associated with disease activity and may preceded relapse
- false positive results are unusual (<3%)

244
Q

speckeld staining pattern in SLE

A
  • associated with antibodies to extractable nuclear antigens (ENA)
  • specificity is for some ribonucleoproteins (e.g., Ro, La, Sm, U1RNP)
  • confirmed with ELISA
  • Ro, La, Sm and U1RNP may occur in SLE
  • Ro and La are characteristically found in Sjogren’s syndrome
  • titres are NOT helpful in monitoring disease activity

other:
- Scl70, RNA polymerase, fibrillarin (may occur in diffuse cutaneous systemic sclerosis)
- Mi2, SRP (may occur in idiopathic inflammatory myopathies)

245
Q

complement activation and profiles in SLE

A
  • formation of antibody-antigen immune complexes will activate complement via the classical pathway
  • complement components become depleted if constantly consumed
  • quantification of C3 and C4 acts as a surrogate marker for disease activity
  • NB: we measure unactivated complement proteins rather than activated ones

SLE:
inactive disease: normal C3, normal C4
active disease: normal C3, low C4
severe active disease: low C3, low C4

246
Q

antiphospholipid syndrome

A

triad of:
- recurrent venous or arterial thrombosis
- recurrent miscarriage
- thrombocytopaenia

may occur alone (primary) or in conjuction with autoimmune disease (secondary)

2 major types of antibody test:
- anti-cardiolipin antibody
- lupus anticoagulant (prolongation of phospholipid-dependent coagulation tests, cannot be assessed if the patient is on anticoagulant therapy)

247
Q

systemic sclerosis pathophysiology

A

inflammation with Th2 and Th17 cells predominating

cytokines lead to activation of fibroblasts and the development of fibrosis
- polymorphisms in type I collagen alpha 2 chains and fibrillin 1 may be important
- polymorphisms in TGF-beta have also been described

cytokines lead to activation of endothelial cells and contribute to microvascular disease
loss of B cell tolerance to nuclear antigens

248
Q

limited cutaneous systemic sclerosis (CREST)

A

skin involvement does NOT progress beyond forearms (although it may involve perioral skin)

characterised by:
- calcinosis
- raynaud’s phenomenon
- esophageal dysmotility
- sclerodactyly
- telangiectasia
(- also primary pulmonary hypertension)

249
Q

diffuse cutaneous systemic sclerosis

A
  • skin involvement does progress beyond the forearms
  • CREST features
  • more extensive GI disease
  • interstitial pulmonary disease
  • scleroderma kidney/renal cysts
250
Q

ANA staining in systemic sclerosis - diffuse vs limited cutaneous systemic sclerosis

A

diffuse cutaneous systemic sclerosis:
- nucleolar pattern
- anti-topoisomerase antibodies (Scl70)
- RNA polymerase
- fibrillarin

limited cutaneous systemic sclerosis:
- anti-centromere antibodies

251
Q

idiopathic inflammatory myopathy

A

dermatomyositis
- within muscle - perivascular CD4 T cells and B cells
- immune complex mediated vasculitis (type III response)

polymyositis
- within muscle - CD8 T cells surround HLA class I expressing myofibres
- CD8 T cells kill myofibres via perforin/granzyme - type IV response

positive ANA (in some patients)
- dermatomyositis: anti-aminoacyl tRNA synthetase antibody (e.g., Jo-1) (cytoplasmic)
- polymyositis: anti-signal recognition peptide antibody (nuclear and cytoplasmic)
- dermatomyositis > polymyositis: anti-Mi2 (nuclear)

252
Q

Chapel Hill classification of systemic vasculitis

A

large vessel:
- Takayasu’s arteritis
- giant cell arteritis/polymyalgia rheumatica

medium vessel:
- polyarteritis nodosa
- Kawasaki disease

small vessel (ANCA associated):
- microscopic polyangiitis
- granulomatosis with polyangiitis
- eosinophilic granulomatosis with polyangiitis

small vessel (immune complex):
- anti-GBM disease
- IgA disease
- cryoglobulinaemia

253
Q

small vessel vasculitis associated with ANCA

A
  • microscopic polyangiitis
  • granulomatosis with polyangiitis
  • Churg-Strauss syndrome (eosinophilic granulomatosis with polyangiitis)
254
Q

pathophysiology of ANCA

A
  • antibodies specific for antigens located in primary granules within cytoplasm of neutrophils
  • inflammation may lead to expression of these antigens on cell surface of neutrophils
  • antibody engagement with cell surface antigens may lead to neutrophil activation (type II hypersensitivity)
  • activated neutrophils with endothelial cells causing damage to vessels (vasculitis(
255
Q

2 types of ANCA

A

cANCA
- cytoplasmic fluorescence
- associated with antibodies to enzyme proteinase 3
- occurs in >90% of patients with granulomatous polyangiitis with renal involvement

pANCA
- perinuclear staining pattern
- associated with antibodies to myeloperoxidase
- less sensitive and specific than cANCA
- associated with microscopic polyangiitis and eosinophilic granulomatosis with polyangiitis

256
Q

mechanism of anaphylaxis

A

type I hypersensitivity response
- cross-linking of IgE on mast cells
- leads to degranulation of mast cells
- this includes the release of biological mediators such as histamines and leukotrienes

results in:
- increased vascular permeability
- smooth muscle contraction
- inflammation and increased mucus production

257
Q

clinical features of anaphylaxis

A
  • feeling of impending doom, loss of consciousness, death
  • angioedema of lips and mucous membranes
  • laryngeal obstruction, stridor
  • hypotension, cardiac arrhythmias, MI
  • vomiting, diarrhoea, abdo pain
  • itch of palms, soles of feet and genitalia
  • wheeze, bronchoconstriction
  • flushing, urticaria
  • conjuctival injection, rhinorrhoea, angioedema

NB: urticaria/angioedema is the MOST COMMON manifestation of anaphylaxis followed by upper airway oedema

258
Q

differential diagnoses for collapse

A
  • MI
  • arrhythmia
  • acute asthma attack
  • PE
  • vasovagal syncope
  • epilepsy
259
Q

immediate treatment of anaphylaxis

A

ABC approach

may require respiratory support
- intubation if severe bronchoconstriction
- tracheostomy if there is severe upper respiratory tract absorption

oxygen by mask
- improves oxygen delivery

IM adrenaline (0.5 mg for adult and may repeat)
- acts on beta-2 adrenergic receptors to constrict arterial smooth muscle
- increased BP, limits vascular leakage, bronchodilator

IV antihistamines (10mg chlorpheniramine)
- opposes the effects of mast cell-derived histamines

nebulised bronchodilators
- assists oxygen delivery

IV corticosteroids (200mg hydrocortisone)
- systemic anti-inflammatory effect
- effects take about 30mins to begin and peak effects take several hours
- important for preventing rebound anaphylaxis

IV fluids
- increases circulating blood volume, thereby increasing blood pressure

260
Q

common causes of anaphylaxis

A

foods:
- peanuts
- tree nuts
- fish and shellfish
- milk
- eggs
- soy products

insect stings
- bee venom
- wasp venom

chemicals, drugs and other foreign proteins
- penicillin/other antibiotics
- IV anaesthetic agents
- latex

261
Q

disorders associated with recurrent meningococcal meningitis

A

Immunological:
1) complement deficiency
- increases risk of infection by encapsulated organisms (meningococcus, pneumococcus, Neisseria gonorrhoeae, H influenzae type B)

2) antibody deficiency
- recurrent bacterial infections
- esp. upper and lower respiratory tract

Neurological:
any disturbance of the blood-brain barrier:
- occult skull fracture
- hydrocephalus

262
Q

investigations for recurrent meningococcal meningitis

A

complement:
- C3, C4
- CH50
- AP50

immunoglobulins:
- serum IgG, IgA, IgM
- protein electrophoresis

CH50
- CH50 is a functional test of the integrity of the classical complement cascade
- all components of the cascade need to be in place for the test to give a positive (normal) result

AP50
- AP50 is a functional test of the integrity of the alternative complement cascade
- all components of the cascade need to be in place for the test to give a positive (normal) result

263
Q

complete deficiency of C7 management

A

vaccinations:
- meningovax
- pneumocax
- Hib vaccine

daily prophylactic penicillin

high level of suspicion

264
Q

summary of complement deficiencies and recurrent meningococcal infections

A
  • any deficiency of the complement pathway may be associated with recurrent meningococcal infection
  • in particular, deficiency of the alternative or final common pathway
  • generally, suggested that adults with sporadic meningococcal disease should be screened for complement deficiency
  • CH50 and AP50
265
Q

serum sickness post penicillin - mechanism

A
  • penicillin can bind 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
  • this means that subsequent exposure to penicillin leads to:
    a) formation of immune complexes with circulating penicillin
  • production of more IgG antibodies
266
Q

consequences of immune complex deposition

A
  • deposition of IgG immune complexes in glomeruli causes renal dysfunction
  • immune complex deposition in joints causes arthralgia
  • immune complex deposition in skin causes vasculitis with local haemorrhage (purpuric)
267
Q

immune complex deposition - deterioration of renal function

A
  • deposition of immune complexes in the glomeruli leads to complement activation and immune cell (macrophage and neutrophil) infiltration
  • this leads to inflammation of the glomeruli (glomerulonephritis)
  • results in increased serum creatinine, proteinuria and haematuria
268
Q

immune complex deposition - disorientation

A

small vessel vasculitis affecting the cerebral blood vessels can compromise oxygen supply to the brain

269
Q

immune complex deposition - purpura

A
  • inflamed blood vessels are likely to leak
  • results in local haemorrhage
  • also becomes plugged with clots, further compromising oxygen delivery
270
Q

X-linked agammaglobulinaemia (+management)

A
  • failure of pre-B cells to mature in the bone marrow
  • this leads to failure to produce immunoglobulin

management: immunoglobulin replacement therapy
- pooled serum immunoglobulin
- administered every 3 weeks
- indefinite treatment

271
Q

mutliple myeloma

A

= a neoplastic proliferation of plasma cells

results in massive expansion of a single plasma cell clone:
- production of excess amounts of a single immunoglobulin molecule with single specificity (usually both heavy and light chain)
- leads to increased numbers of abnormal plasma cells in the BM
- causes lytic bone lesions

  • unusual or vertebral fracrures
  • anaemia
  • high ESR
  • high calcium
  • measure immunoglobulins/electrophoretic strip
  • urinary Bence-Jones proteins
  • skeletal survey
  • refer to haematologist for specialist management
272
Q

management of rheumatoid arthritis

A

1st line
- disease modifying drugs (e.g., methotrexate)
- sulphasalazine, hydroxychloroquine and leflunomide are also frequently used in addition or if methotrexate is not tolerated

further treatment:
- TNF-alpha antagonists (inhibits downstream inflammation)
- rituximab (anti-CD20 which depleted mature B cells but not plasma cells)
- abatacept (CTLA4-Ig fusion protein binds to CD80 and CD86 and inhibits T cell activation)
- tocilizumab (antibody against IL6 receptor)