Immune System Flashcards

1
Q

What is the immune system?

A

A complex network of cells and soluble molecules which interact with one another to remove foreign material from the body. Includes bacteria, viruses, parasites and other infection-causing pathogens, as well as some cancer cells.

Extremely effect in performing its function but error can occur in this highly complex system, which results in allergic reaction, immunopathology or autoimmunity.

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

Name 4 importances of the immune system.

A
  • Immune system plays an important role in inflammation and immunity.
  • Immune system failure can cause severe disease, such as primary immune deficiencies and immunodeficiency viruses.
  • Many diseases or symptoms are caused by the immune system: allergy, autoimmunity and immunopathology.
  • Understanding how to harness the power of the immune system can be extremely beneficial to vaccination and immunotherapy for example.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Use feline influenza virus as an example of when the immune system fails to control disease.

A
  1. FIV is characterised by a decline of CD4 T-cells
  2. T-cell immunodeficiency results in symptoms
  3. CD8 T-cells control viral load during chronic phase
  4. This control does not last forever
  5. CD8 T-cells get exhausted
  6. Virus escapes from the CD8 T-cell response
  7. Highlights the importance of T-cell immunity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Define immunopathology.

A

Tissue damage caused by an excessive immune response.

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

Define autoimmunity.

A

The failure to distinguish self from non-self.

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

Define allergy.

A

Inappropriate response to an environmental antigen.

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

What is the aim of vaccination?

A

Vaccination aims to establish immunological memory.

  1. Primary immune response
  2. On exposure to the pathogen, secondary immune response because the immune system has immunological memory to get a rapid and robust response.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Where do all cells in the immune system arise from?

A

Pluripotent haematopoietic stem cell from bone marrow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Name the cells that these stem cells give rise to.

A
  • Common lymphoid progenitor cells
  • Common myeloid progenitor cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What do common lymphoid progenitor cells give rise to?

A
  • B cells – when activated, they differentiate into plasma B cells, which produce antibodies.
  • T cells – activate to become activated T cells
  • NK cells – natural killer cells, important for viral infection control
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What do common myeloid progenitor cells give rise to?

A

Granulocyte/macrophage progenitor cells and megakaryocyte/erythrocyte progenitor cells

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

What do megakaryocyte/erythrocyte progenitor cells give rise to?

A

Platelets and red blood cells

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

What do granulocyte/macrophage progenitor cells give rise to?

A
  • Neutrophils
  • Eosinophils
  • Basophils
  • Unknown precursor of mast cell and monocytes, which differentiate to form macrophages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What do dendritic cells do?

A

Forms a link between the innate and adaptive immune system

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

What are the cellular components of innate immunity?

A

Phagocytes
Macrophages/monocytes
Neutrophils
Eosinophils
Basophils
Mast cells
Natural killer cells

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

What are the characteristics of innate immunity?

A
  • Recognition of the nature of the challenge, for example, recognition is non-specific
  • No memory of the challenge, as the second encounter triggers the same response
  • Barriers to infection
  • Soluble factors involved in immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What is the function of innate immunity at best and worst?

A

At best: can eradicate infection

At worst: slows down/delays infection until adaptive immune response is generated

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

Name and describe 5 barriers.

A
  • Skin – physical barrier, chemical barrier (sweat) and microbial barrier (commensals)
  • Respiratory tract – patrolled by alveolar macrophages and mucociliary escalator
  • Alimentary tract – physical barrier (peristalsis) and chemical barrier (stomach acid)
  • Urinary tract – low pH of urine and flushing
  • Corea/conjunctiva – blinking and antibodies in tears
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Name and give the function of the cells of the innate immune system.

A
  • Macrophages – kill intracellular pathogens
  • Sentinel cells – raise alarm following infection
  • Neutrophils – kill rapidly dividing bacteria
  • Eosinophils – kill parasites
  • NK cells – kill virus infected cells
  • Mast cells – trigger inflammatory response
  • Dendritic cells – activate adaptive immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are the 3 functions of the soluble factors of the innate immune system?

A
  • Complement – coats/opsonises pathogens with layer of molecules that cells of the innate immune system have receptors for/complement receptors.
  • Acute phase – similar to complement proteins. Also activates complement system.
  • Interferons – activate cells to produce anti-viral proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Which cells are capable of phagocytosis?

A

Mononucleated leukocytes are capable of phagocytosis: monocytes and macrophages
Polymorphonucleated leukocytes are also capable of phagocytosis: neutrophils, basophils and eosinophils

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

Give a brief overview of phagocytosis.

A
  1. Chemotaxis
  2. Adherence
  3. Membrane activation
  4. Initiation of phagocytosis
  5. Phagosome formation
  6. Fusion
  7. Killing and digestion
  8. Release of degradation products
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Define antigen.

A

A substance which binds to a lymphocyte receptor and in so doing may initiate an immune response.

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

Name the 4 types of antigen.

A

Heteroantigens
Autoantigens
Alloantigens
Xenoantigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What are heteroantigens?

A

Foreign to the body: infectious agents, environmental substances and chemicals like drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are autoantigens?

A

Also called self-antigens, recognition triggers autoimmunity

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

What are alloantigens?

A

Cells from same species but genetically different

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

What are xenoantigens?

A

Cells from a different species. Particular interest for transplantation field.

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

Describe the concept of epitopes and immunodominance.

A

Antigenic epitope (determinant) – one antigen may have multiple epitopes, some of which may be immunodominant – majority of immune response is targeted at a certain set of epitopes.

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

What is a hapten?

A

A small chemical group, which when chemically conjugated to a large molecules (carrier protein) will alter its specificity creating a new epitope. A hapten cannot by itself stimulate antibody production, but can combine with antibody once linked to a carrier protein.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What is the clinical relevance of haptens?

A

Cutaneous drug eruption: drug (ketoconazole) binds to a dermal protein which stimulates an Immune response.

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

What is an adjuvant?

A

A substance which, when combined with antigen, non-specifically enhances the immune response. May also have a depot effect allowing slow release of antigen.

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

Describe the structure of an immunoglobulin.

A

2 heavy and 2 light chains, bound by disulphide bridges.
At the end of each, there are domains labelled V, meaning variable. Variability is all located at ends, as this determines which antigen the molecule will bind to. Letter C means constant, domains which are the same across molecules.

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

How are immunoglobulins classed?

A

By the heavy chain:
- Gamma = IgG
- Mu = IgM
- Alpha = IgA
- Delta = IgD
- Epsilon = IgE

Each with a kappa or lambda light chain.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Define valence.

A

Number of antigens an antibody can bind to

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

What is the structure of IgG?

A

Single Y shaped unit, gamma heavy chain, valance 2, subclasses exist

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Where is IgG found?

A

Main serum Ig, for example, found in the blood. Diffuses easily to extravascular tissue.

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

What are the functions of IgG?

A
  • Fixes complement
  • Opsonin
  • Neutralise toxins, such as bacterial toxins
  • Main Ig in secondary immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

What is opsonin?

A

Enhancing phagocytosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

What is the structure of IgM?

A
  • 5 Y shaped units
  • Valence 10
  • Mu heavy chain
  • Extra heavy chain domain (CH4)
  • J chain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Where can IgM be found?

A

Intravascular, stays in the blood due to large size

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

What are the functions of IgM?

A
  • Agglutinates particulate antigen
  • Fixes complement
  • Main Ig in early immune response
  • Important in bacteraemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What is the structure of IgA?

A
  • Valence 4
  • Monomer or dimer in serum (species dependent)
  • Dimer in mucosal secretions (all species)
  • Alpha heavy chain
  • Dimer linked by J chain
  • Subclasses exist
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What are the functions of IgA?

A
  • Dimer at mucosal surfaces has secretory piece – provides protection from enzymatic degradation
  • Mucosal immune defence
  • Inhibits microbial attachment
  • Neutralises toxin
  • Weak opsonin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What is the structure and location of IgD?

A
  • 1 Y shaped unit
  • Valance 2
  • Delta heavy chain
  • Found only on surface of immature B lymphocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What is the structure of IgE?

A
  • 1 unit, valence 2
  • Epsilon heavy chain
  • Extra domain CH4
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Where is IgE found?

A
  • Low level in serum
  • More in animals than man
  • Most bound to tissue mast cells or blood basophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What are the functions of IgE?

A
  • Involved in parasite rejection
  • Involved in allergy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

How do antigens and antibodies interact?

A

The antigenic epitope slots into a groove formed between the hypervariable regions of the heavy and light chains. Specific attachment – lock and key.

Firm attachment – electrostatic forces, hydrogen bonds, hydrophobic forces and van der waal’s forces. Affinity – binding strength of antibody for its epitope.

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

Distinguish primary and secondary lymphoid tissue.

A

Primary – where the immune system develops.

Secondary – site of immune responses.

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

Name the 4 primary lymphoid tissues.

A

Bone marrow
Ileal peyers patch
Thymus
Bursa of fabricius in birds

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

Describe the role of bone marrow in the immune system.

A
  • Stem call – pluripotent
  • Origin of T and B lymphocytes
  • Site of maturation of B lymphocytes before export to secondary lymphoid tissues in humans, rabbits and rodents
  • Site of some B cell differentiation to plasma cells for antibody production in the humoral response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Describe the role of ileal peyers patch in the immune system.

A
  • Site of B cell maturation in most animal species
  • Found in the small intestine, domes without villi
  • Secondary follicle with germinal centres are where T cells are
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Describe the role of bursa of fabricius in the immune system.

A
  • Site of B cell maturation in birds
  • Lymphoepithelial organ near cloaca

Bursectomised chicks cannot make humoral immune response/cannot make antibodies

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

Describe the role of the thymus in the immune response.

A

Anterior mediastinum in front of the heart. Involutes with age. Thymectomised or congenitally athymic (‘nude’) animals cannot mount cell-mediated immune responses.

  • Site of extramedullary T cell development – takes place outside bone marrow.
  • Cells generate cell mediated immunity.
  • Humans, rabbits and rodents have B cell maturation before circulation.
  • T cells arise in bone marrow and in all species migrate to thymus to complete maturation and then are circulated.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What are the 2 types of secondary lymphoid tissue?

A

Encapsulated – spleen and lymph nodes

Unencapsulated – mucosal lymphoid aggregates

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

Describe the structure of a lymph node.

A
  • Encapsulated so has a capsule and then a subcapsular sinus below.
  • Afferent lymphatics is where immune cells arrive at lymph nodes.
  • Lymph nodes contain immune cells that perforate through the cell and exit via the efferent lymphatics.
  • B and T cells sit in cortex.
  • Primary follicles contain B cells that are inactive that are activated on immune response.
  • Paracortex is where T cells are between follicles.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Describe the structure of the spleen.

A

Encapsulated: trabecula skeleton creates framework and there are areas of red pulp in between containing red blood cells, and areas of whit pulp where lymphocytes/B cells are. PALS, per-arteriolar lymphoid sheath is where T cells are found.

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

Describe secondary unencapsulated lymphoid tissue (MALT) and where they are located.

A

Secondary unencapsulated lymphoid tissue MALT – at the mucosal surfaces which are in contact with the environment.

  • Gastrointestinal (GALT)
  • Bronchial (BALT)
  • Nasal (NALT)
  • Conjunctival (CALT)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

Describe the morphology of small lymphocytes (T and B cells).

A
  • Virgin or memory (inactive)
  • Little cytoplasm (large nucleus)
  • Few organelles
  • Condensed chromatin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Describe lymphoblast (T and B cells) morphology.

A
  • Stimulated by antigen
  • More cytoplasm
  • More organelles
  • Less condensed chromatin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Describe plasma cell/late stage B cell morphology.

A
  • More endoplasmic reticulum
  • Clock face chromatin
  • Antibody production and secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

How can B and T cells be distinguished?

A
  1. Anatomical location
  2. Function
  3. Expression of surface molecules unique to each population
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is the process of immunofluorescence?

A

Expression of these molecules by individual cells in suspension or tissue can be determined using antibodies specific for the molecule of interest which are conjugated to a fluorochrome.

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

Describe the process of immunofluorescence.

A
  1. Antibody to SmIg conjugated to fluorochrome.
  2. Added to unknown lymphocytes.
  3. Antiserum labels SmIg in binding.
  4. Expose to UV light to excite the fluorochrome.
  5. Examine microscopically or count with a flow cytometer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What are flow cytometers used for?

A

Used both clinically in labs and by immunologists in research labs:

  • Can take a single sample, stain antibodies, and begin looking at different immune cells and analyse their phenotypes.
  • Enables simultaneous measurement of up to 50 different characteristics of a single cell.
  • Identification and analysis of distinctive phenotypes in heterogenous populations.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Why do lymphocytes recirculate?

A

Lymphocytes recirculate to maximise the chance of contact between an antigen and the appropriate responding populations of lymphocytes.

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

Give the path of lymphocyte recirculation.

A
  1. Tissue
  2. Afferent lymphatics
  3. Lymph nodes
  4. Efferent lymphatics
  5. Thoracic duct
  6. Bloodstream
  7. Can exit into required areas and tissues via HEVs (high endothelial venules)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is lymphocyte homing?

A

Interaction between vascular addressins and homing receptors in the HEVs triggers diapedesis and turbulence, which increases the chance of interactions. Located in areas where lymphocytes exit tissues.

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

Give an example of specialised recirculation.

A

The common mucosal system: cells activated in response to an event at 1 mucosal surface can home to other mucosal surfaces.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is the importance of the common mucosal system?

A

Generation of colostral antibody

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

Define complement.

A

A series of plasma proteins (around 30) which when activated, interact sequentially forming a self-assembling enzymatic cascade generating biologically active molecules mediating a range of end processes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What are the distinct phases of the complement system which result in the elimination of microbes?

A
  1. Pattern recognition
  2. Protease (enzymatic) cascade amplification and C3 and C5 convertase production
  3. Inflammation
  4. Enhanced phagocytosis/opsonisation
  5. Membrane attack – lysis of microbial membranes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What happens once a complement pathway is triggered?

A

Once triggered, pathways get enzymatic cascades, each with different components of the cascade and all 3 result in the production of C3 convertase and C5 convertase.

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

What does the classical complement pathway involve?

A

Triggered by complexes between antigens and antibodies. For classical pathway, must have already had an immune response to produce antibodies. Antigen antibody complexes on the surfaces of pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Outline the process of the classical complement pathway.

A
  1. Triggered by antigen-antibody complexes on a pathogen surface. Antibodies specific to antigen from previous immune response.
  2. Binding of the first component, C1, which binds to part of the antibody at the CH2 region for complement fixation.
  3. C1 is made up of several units and there is sequential activation of each component.
  4. Active C1s component acts on the 2nd component, C4, causing C4 to split into 2 parts, a and b.
  5. C4b becomes attached to bacterial surface and C4a stays in the serum.
  6. C1s also acts on 3rd component, C2, which splits C2 into 2 parts, a and b.
  7. C4b and C2a attach to each other on the pathogen surface and together they form the C3 convertase.
  8. C3 convertase acts on C3, casing C3 to split into 2 components, a and b.
  9. C3a stays in the serum and C3b goes to the pathogen surface.
  10. C4b, C2a and C3b attach together on the pathogen surface and together form the C5 convertase.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What does the MB-lectin complement pathway involve?

A
  1. Mannose-binding lectin binds mannose on pathogen surface
  2. MBL, MASP-1, MASP-2, C4, C2
  3. C3 convertase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What does the alternative complement pathway involve?

A
  1. Triggered by pathway surfaces
  2. C3, B, D
  3. C3 convertase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What are the functions of C3a and C5a?

A

Peptide mediators of inflammation and phagocyte recruitment.

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

What are the functions of C3b?

A

Binds to complement receptors on phagocyte for opsonisation of pathogens and removal of immune complexes.

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

Name the terminal complement components and their function.

A

C5b
C6
C7
C8
C9
Membrane attack complex and lysis of certain pathogens and cells.

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

What is the terminal pathway?

A

Where C5 convertase breaks down C5 and results in the membrane attack complex, which causes lysis of pathogens (occasionally cells).

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

What are the consequences of complement activation?

A

Inflammation – due to the generation of bioactive substances, such as anaphylatoxins and chemoattractants.

Removal of particulate antigens and immune complexes – enhanced phagocytosis.

Cytolysis

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

How do C3a and C5a cause inflammation?

A
  • Smooth muscle contraction
  • Mast cell degradation
  • Vasodilation, local oedema, local influx of antibody and complement, cell extravasation
  • Neutrophil activation
  • Attracted to chemotactic gradient
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What 3 ways is phagocytosis enhanced?

A
  1. Phagocytosis by non-specific adherence - IgM increases agglutinates particulate antigens and so there is a greater chance of interaction.
  2. Opsonisation - specific binding between phagocyte receptors and pathogen antigens, as they are complementary. Complement involves as well further opsonisation.
  3. Removal of immune complexes - antigen coated in C3b attached to red blood cell as they have receptors and circulate around in the blood attached. When supplying liver or spleen, antigens are removed.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What is cytolysis?

A
  • C5 split into C5a and C5b
  • C5b starts to sequentially assemble itself with final components of complement pathway, C6, 7, 8 and 9. Forms circular structure.
  • Circular structures are inserted into pathogen membranes and causes influxes of water and osmotic lysis.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

How is the complement cascade controlled?

A

Normal cells have membrane proteins which breakdown C3 convertase. This protects normal cells. For example. DAF, decay-accelerating factor.

CD59 expressed widely on membranes and prevents formation of the MAC on normal cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What are the consequences of a C3 deficiency?

A

Causes susceptibility to bacterial infections and get symptoms from prolonged immune response. High temperatures and recurrent infections could be signs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What is the concept of a complotype?

A

An individual might have multiple polymorphisms that causes susceptibility to certain infectious diseases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

Why are T cells important?

A
  • Control/eliminate infection
  • Immunity and response to vaccination
  • Kill cancerous cells
  • Disease pathogenesis – transplant rejection, autoimmune disease, leukaemia/lymphoma
  • Therapeutic applications – cellular therapy against cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What are the different functional classes of T cells

A

CD8+ T cells – killer T cells

CD4+ T cells / TH / T helper cells
- TH1
- TH2
- TH17
- TFH (follicular helper)

CD4+ T regulatory cells – rather than stimulate an immune response, they dampen immune responses. In theory, could be used to treat autoimmune diseases.

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

What do T cells recognise?

A

T cells cannot recognise complex or whole antigens. T cells recognise small peptide fragments bound to Major Histocompatibility Complex molecules which are expressed at the cell surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What is the role of Major Histocompatibility Complexes?

A

T cells express T cell receptors which are responsible for recognising cell surface peptide MHC. Therefore to trigger a T cell response, complex antigens must first be converted into a user friendly form.

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

Define antigen processing and presentation.

A

Refers to the generation of small peptide fragments which are bound to major histocompatibility complexes and transported to the cell surface.

  1. Antigen uptake
  2. Antigen processing
  3. Antigen presenting at the cell surface
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Describe the class I MHC processing pathway.

A

CD8+ T cells see T cell epitopes presented by class I MHC, found on most nucleated cells:

  1. Antigens found in the cytoplasm, viral, self or tumour, are referred to as endogenous or cytosolic antigens.
  2. Endogenous (cytosolic) antigens are processed and presented with MHC class I molecules
  3. Presented peptide fragments are usually 8-14 amino acids in length.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

What is the role of MHC class I in viral protein synthesis?

A
  1. Peptide fragments of viral proteins are transported into the endoplasmic reticulum
  2. Peptide fragments are bound by MHC class I and delivered to the cell surface
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are the important components of MHC class I processing pathway?

A
  • The proteasome generates peptide fragments from cytosolic protein. During an immune response induced by interferon, modifications to the proteasome mean that it produces peptides that bind well to MHC class I.
  • TAP, Transporter associated with antigen processing, delivers peptides to the endoplasmic reticulum.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Where are MHC class II found?

A

CD4+ helper T cells see T cell epitopes presented by class II MHC.
MHC class II is found on antigen presenting cells: dendritic cells, macrophages, B cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe the MHC class II processing pathway.

A
  1. Antigens taken up from outside the APC, extracellular pathogens and toxins, are referred to as exogenous antigen.
  2. Exogenous antigens are processed and presented by MHC class II at the cell surface.
  3. MHC class II molecules are found on specialised antigen presenting cells – dendritic cells, macrophages, B lymphocytes.
  4. Presented peptide fragments are usually 15-24 amino acids in length.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What are the professional antigen presenting cells that express MHC class II?

A

Dendritic cells – several different types
Macrophages
B-cells

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

What are non-professional antigen presenting cells?

A

A range of cells hat can be induced to express MHC class II and present antigen when exposed to IFN-gamma.

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

What are the functions of dendritic cells?

A
  • Activate naïve T cells, highly potent APCs
  • Large surface area to increase efficiency of antigen uptake
  • Widespread distribution – most epithelia and solid organs, such as the heart and kidneys
  • Phagocytosis – actively ingest antigens using complement receptors and Fc receptors
  • Micropinocytosis – engulf large amounts of surrounding fluid non-specifically.
  • Use pattern recognition receptors to recognise pathogen.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

How do dendritic cells activate naive T cells?

A
  1. Dendritic cells take up microbial antigens and become activated in peripheral tissues.
  2. Activated dendritic cells travel to the lymph nodes.
  3. At the lymph nodes, the dendritic cells become mature and activate T cells.
  4. CD maturation involves increased expression of MHC molecules, express co-stimulatory molecules and secrete cytokines. All 3 endow dendritic cells with a potent ability to activate naïve T cells.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What are pattern recognition receptors?

A
  • Recognise highly conserved microbial components referred to as pathogen associated molecular patterns or associated molecules patterns.
  • Sugars, proteins, lipids, nucleic acid motifs
  • Not easily altered by microbes to avoid detection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What are cytokines?

A

Soluble factors released by one cell that bind to specific receptors on the same cell or another cell leading to altered function. Important roles in all aspects of the immune response.

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

What are the properties of cytokines?

A
  • Produced by many cell, and 1 cell can make many cytokines
  • Low MW proteins
  • Produced locally and transiently
  • Potent at picomolar concentrations
  • Have specific receptors
  • Binding alters target cell function
  • May have multiple actions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What are the 4 different roles cytokines can have in the immune system?

A
  1. Haemopoietic, for example GM-CSF
  2. Regulatory, for example IL-10
  3. Cytotoxic, for example IFN
  4. Autocrine, for example IL-2
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Define a chemokine.

A

Chemotactic recruitment of cells from blood into tissue.

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

Describe feline mammary carcinoma as an example of an antigen processing defect.

A
  • Defects in antigen processing are common in human tumours
  • Feline mammary carcinomas – reduced expression of MHC class I HC and proteosome components (LMP2 and LMP7) compared to normal mammary tissue.
  • May cause defective processing of some tumour antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Describe viruses that produce UL49.5 proteins as an example of an antigen processing defect.

A
  • Bovine herpesvirus 1 (BoHV-1), pseudorabies virus and equid herpesviruses 1 and 4 encode a UL49.5 protein that inhibits TAP mediated peptide transport.
  • This impairs peptide loading of MHC class I molecules and represents an immune evasion mechanism.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is MHC restriction?

A

Only activate at the right combination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What features of the MHC make life hard for pathogens?

A
  • Polygenic: contains several different copies MHCI and MHCII genes and all these genes are expressed at the same time.
  • Polymorphic: multiple variants (alleles) of each gene in the population
  • Every individual expresses a number of different MHC antigens. The specific set of MHC expressed by an individual is referred to as “tissue type”. Each set of MHC will be capable of presenting a different range of peptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What are the clinical implications of MHC?

A
  • Viral infection that affects a group of animals with different tissue types
  • Impact on being able to find transplant donors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Describe the inheritance of MHC.

A
  • Every individual has two sets of MHC alleles called an “MHC Haplotype”
  • MHC gene expression is co-dominant
  • Greater diversity is possible with MHCII genes as different α and β chains may pair together to give distinct MHCII products
  • Fundamentally Mendelian - 1/4 chance of a sibling being a matched donor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Distinguish the human and chicken MHC.

A
  • Chicken MHC is 1/20th of the size of the human MHC at only 19 genes
  • Chicken MHC has only a single classical class I and class II molecule expressed at high level
  • Single dominantly expressed MHCI and MHCII molecules confer resistance or susceptibility to particular pathogens (leading to strong genetic associations with infectious disease)
  • Chickens may live or die according to their dominantly expressed class I molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Name the 3 significances of MHC to veterinary medicine.

A
  1. Graft Rejection
  2. Associated with disease susceptibility, such as increasing the risk of developing a disease, especially autoimmune disease.
  3. Association with disease resistance and production traits in livestock.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

Name 4 types of graft.

A
  • Autografts: same individual
  • Isograft: genetically identical
  • Xenograft: different species (pigs)
  • Allograft: same species, different genetic constitution (most common)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What is alloreactivity?

A
  • Alloreactivity is the recognition of peptides complexed with non-self MHC
  • It is estimated that 1 – 10% of all T-cells have the potential to be alloreactive which is why graft rejection can be so destructive.
  • Graft rejection may be acute (days to weeks) or chronic (months to years)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

List canine diseases with clear MHC associations.

A

Canine rheumatoid arthritis
Immune-mediated haemolytic anaemia (IMHA)
Diabetes mellitus
Lymphocytic thyroiditis
Addison’s disease
Chronic hepatitis in Dobermans
Necrotizing meningoencephalitis
Anal furunculosis
Chronic superficial keratitis
Susceptibility to leishmaniosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

Define an anti-inflammatory reaction.

A

Biological response to harmful stimuli aimed at removing that stimuli and promoting healing. Should be a local response.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

Give 4 examples of when anti-inflammatory responses go wrong.

A
  • Type 1 reactions/anaphylaxis, such as vaccination reactions
  • Airway disorder, such as feline asthma or recurrent airway obstruction in horses
  • Autoimmune haemolytic anaemia in dogs
  • Sweet itch in horses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are the main anti-inflammatory vascular responses?

A

Vasodilation – caused by many mediators, such as histamine and prostaglandins

Increased vascular permeability – caused by many mediators, such as histamine

Exudation – fluid filters from the circulatory system into surrounding tissue. Carries leukocytes and components to proteolytic enzyme cascade, such as complement, coagulation, fibrinolytic and kinin systems.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What are the principal mediators of inflammation and allergy?

A
  • Histamine
  • Bradykinin
  • Nitric oxide
  • Cytokines – interleukins, interferons, chemokines, colony stimulating factors
  • Eicosanoids – prostaglandins, thromboxanes, leukotrienes
  • Other plasma proteins – complement, coagulation and fibrinolytic factors
  • Platelet activating factor
  • Neuropeptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

Where do the principal mediators of inflammation and allergy act?

A

autocoids/local hormones. Released from cells upon stimulation.

  • Act on adjacent/local cells – paracrine
  • Act on cell that secreted hormone – autocrine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Why must histamine be deactivated and how?

A

Keep them acting locally, done by 2 different pathways:

  1. Diamine oxidase
  2. Histamine-N-methyl-transferase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What are the 5 roles of histamine?

A
  • Tissue repair, inflammation
  • Control of local blood supply
  • Contributes to allergic and anaphylactic reactions
  • Neurotransmission in the CNS
  • Gastric acid secretion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Where is histamine found throughout the body?

A
  • High concentrations in the lungs, skin, GI tract and brain
  • Present in mast cells and basophils – contain granules densely packed with histamine, heparin, proteoglycans and serine proteases in a ratio of 1:3:6
  • Neurones in the brain
  • Enterochromaffin like cells in the stomach
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Where are mast cells produced and found?

A
  • Produced by bone marrow
  • Released as immature cells and then mature in tissue
  • Found beneath skin throughout the respiratory, digestive and urinary tracts.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What is mast cell degranulation?

A
  1. Priming of mast cell: exposure to allergen creates IgE antibodies, which bind to mast cell IgE receptors.
  2. 2nd exposure to allergen caused allergen to bind to IgE on mast cell surface.
  3. Activates mast cell by: increasing intracellular calcium concentration or C3a and C5a complement components
  4. Leads to degranulation and histamine release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

How is histamine release inhibited?

A

Increase in cAMP by beta-adrenoreceptor agonists

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What is the effect of H1 receptor activation?

A

Systemic vasodilation
Increased vascular permeability
Itching
Bronchoconstriction
Ileum contraction
Effects on neuronal action potential firing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is the receptor coupling for H1 receptors?

A

Increased IP3 and DAG, stimulating calcium ion release.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What is the effect of H2 receptor activity?

A

Stimulate gastric acid secretion
Relax smooth muscle
Speed up heart rate
Inhibit antibody and cytokine production
Inhibit neutrophil activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What is the receptor coupling of H2 receptors?

A

Increased cAMP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What is effect of H3 receptor activation?

A

Inhibits neurotransmitter release from neurones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is the receptor coupling for H3 receptors?

A

Decreases cAMP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is the effect of H4 receptor activation?

A

Regulates neutrophil release from bone marrow and chemotaxis in mast cells.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

What is the receptor coupling of H4 receptors?

A

Decreases cAMP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are H1 receptor antagonists?

A
  • Used in treatment of allergy, such as hay fever and urticaria.
  • Orally active
  • Hepatic metabolism
  • There are no H1 antagonists licenced for vet use in the UK. Human drugs given with owner consent.
  • Have limited use in treating allergic disorders in animals.
  • Most useful if given before allergen challenge: feline asthma, allergic skin disorders relief of itching, motion sickness and mild sedation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are 1st and 2nd generation H1 receptor antagonists?

A

1st generation: average duration of 4-6 hours and crosses the blood brain barrier. Such as diphenhydramine, chlorphenamine, cyproheptadine and alimemazine.

2nd generation: longer duration of 12-24 hours and does not cross the blood brain barrier. Such as cetirizine, loratadine and acrivastine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are H2 receptor anatgonists?

A

Inhibit gastric acid secretion.
Treatment of gastric ulcers. For example, cimetidine, ranitidine, famotidine.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What are H3 and H4 receptor antagonists?

A

Used as research tools and have been human clinical trials on antagonists of H3 and H4 receptors.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Define type 1 hypersensitivity reactions.

A

Immediate or anaphylatc hypersensitivity – allergic reaction. Oversensitive reaction to an allergen producing an excessive inflammatory response.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

What is anaphylaxis?

A
  • Whole body reaction of a previously sensitised animal to the sensitising allergen.
  • Life threatening
  • Produces immediate smooth muscle contraction, vasodilation, increased vascular permeability
  • Within 2 minutes, there is nausea, abdominal pain, palpitation, urticaria, difficulty in breathing, hypotension, inadequate circulation.
  • Within 4 minutes there is shock.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What are 3 possible drug treatments for anaphylaxis?

A

Epinephrine – immediate and subcutaneously. Intravenous administration also required to maintain intravascular volume and supply additional; medication.

Chlorphenamine and promethazine – H1 receptor antagonists intravenously

Glucocorticoids – no acute effect but suppress slow onset urticaria, bronchospasm, laryngeal oedema and hypotension.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What are cytokines?

A

Protein or polypeptides produced and released from immune cells during inflammation. Upregulated during inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What are the characteristics of cytokines?

A
  • Very potent – sub nanomolar (< 1 x 10-9 M)
  • More than 100 exist and they are split into 4 main groups: interleukins, interferons, chemokines and colony stimulating factors
  • Very complex – interact with each other, stimulate receptor upregulation for others
  • Act at kinase-linked receptors or GPCRs
  • Coordinate inflammatory response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What are eicosanoids?

A
  • Generated from phospholipids
  • Precursors are 20 carbon fatty acids – eicosa = 20 C
  • They are not stored but generated on demand
  • They are prostaglandins, leukotrienes and thromboxane
  • Involved in many physiological processes with major involvement in inflammation
  • Target of several groups of drugs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

Describe the nomenclature of prostaglandins.

A

PG = prostaglandin
Letter = basic structure
Number = number of carbon C=C double bonds

PGD2, PGE2, PGF2alpha, PGI2

If arachnoid acid is precursor then 2 C=C bonds are present. If diet is rich is omega-3 fatty acids, then the precursor is slightly different to arachnoid acid. PGE3 and PGI3 have more of an inflammatory effect.

150
Q

Describe the enzyme cascade that begins with arachidonic acid.

A
  1. Arachidonic acid to PGG2 and PGH2 by cyclooxygenase.
  2. PGH2 into PGI2, PGF2, PGD2, PGE2 and thromboxanes.
151
Q

What are the 2 isoforms of cyclooxygenase?

A

1 – constitutively expressed in most cells

2 – not normally present, expression induced inflammatory factors by cytokines for example.

152
Q

Describe the effects of PGD2.

A
  • Relaxation of vascular smooth muscles and constriction at high concentrations.
  • Constriction of bronchial muscle
  • Relaxation of GI and uterine muscle
153
Q

Describe the effects of PGE2.

A
  • Relaxation of vascular smooth muscle
  • Dilation and sometimes constriction of bronchial muscles
  • Hyperalgesia
  • Produces fever
  • Promotes platelet aggregation
  • Inhibition of gastric acid secretion
  • Increases gastric mucous secretion
154
Q

What is hyperalgesia?

A

Lower nociceptor threshold level. Sensitises receptors on afferent nerves to agents not usually causing pain.

155
Q

Describe the effects of PGF2alpha.

A
  • Constriction of vascular smooth muscle
  • Constriction of bronchial muscle in dogs and cats
  • Luteolysis in some species, such as cattle, and uterus contraction
156
Q

Describe the effects of PGI2 (prostacyclin).

A
  • Relaxation (potent) of vascular smooth muscle
  • Hyperalgesia
  • Inhibit platelet aggregation
  • Modulates kidney function
157
Q

Where are thromboxanes mostly found?

A

Platelets

158
Q

What are leukotrienes?

A

Leukotrienes – produced in leukocytes, lung, mast cells and platelets. Triene of double bonds.

159
Q

What are the 2 receptor classes for leukotrienes?

A

BLT for LTB4 – causes activation and targeting of leukocytes and cytokine production

Cysteinyl-LTs – cause bronchoconstriction and vasodilation, coronary vasoconstriction and is particularly important in asthma

160
Q

Describe histamines source and main effect in inflammation.

A

Mast cells, basophils

Vasodilation and increased vascular permeability

161
Q

Describe Prostaglandins PGe2 and PGI2’s main source and main effect in inflammation.

A

Mast cells, leukocytes

Vasodilation, pain, fever

162
Q

Describe leukotrienes main source and main effect in inflammation.

A

Mast cells, leukocytes

Increased vascular permeability, leukocyte activation and chemotaxis

163
Q

Describe thromboxane A2’s main source and main effect in inflammation.

A

Platelets

Platelet aggregation and vasoconstriction

164
Q

What are the main inflammatory agents?

A

Non-steroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids

H1 receptor antagonists and chondroprotective drugs

165
Q

What are corticosteroids and how they are produced?

A
  • Steroids secreted by adrenal gland
  • Outer layer of cortex/zona glomerulosa – secreted mineralocorticoids, such as aldosterone, regulate water and electrolyte balance
  • Middle layer/zona fasciculata – secretes mixture of glucocorticoids
  • Main glucocorticoids in humans is hydrocortisone/cortisol and in many animals, the main glucocorticoid is corticosterone
166
Q

How are glucocorticoids released?

A
  • Glucocorticoids released in pulsatile fashion, circadian rhythm, in humans highest in morning, declining to low in evening/night.
  • Produced from cholesterol, initial step regulated by ACTH
167
Q

Describe the action of glucocorticoids.

A
  • Glucocorticoids bind to receptor in the cell cytoplasm
  • Glucocorticoid-receptor complex moves to nucleus
  • Increases or decreases gene transcription
168
Q

List the widespread metabolic effects of glucocorticoids.

A
  • Decreased uptake and utilisation
  • Increased gluconeogenesis
  • Decreased protein synthesis
  • Increased protein catabolism
  • Redistribution of fat
  • Decreased absorption in GI tract
  • Increased excretion from kidney
169
Q

What are the consequences of a loss of corticosteroids?

A

Produces muscle weakness, hypotension, hypoglycaemia and weight loss.

170
Q

What is Addison’s disease?

A

May be autoimmune condition or caused by destruction of adrenal glands by chronic inflammation.

171
Q

What is Cushing’s syndrome? What are its symptoms?

A

Excess of corticosteroids can be caused by prolonged glucocorticoid administration or excessive activity of adrenal glands.

  • Increased, excessive appetite
  • Distended abdomen
  • Coat problems – baldness, hair loss, hair thinning, discolouration
  • Polyuria and polydipsia
172
Q

What is caused by excessive glucocorticoid action?

A
  • Hyperglycaemia – decreased glucose uptake and utilisation and increased gluconeogenesis
  • Muscle wasting – decreased protein synthesis and increased protein catabolism
  • Osteoporosis – decreased calcium absorption in GI tract and increased calcium excretion from kidney
173
Q

What are the anti-inflammatory actions of glucocorticoids?

A
  • Decreased movement of neutrophils from blood vessels
  • Decreased transcription of cytokine and cell adhesion factor genes
  • Decreased activation of mast cells, neutrophils, macrophages
  • Decreased activation of T helper cells
  • Decreased fibroblast function – reduced healing and repair
  • Decreased osteoblast (build bone) activity
  • Decreased transcription of many ILs, TNF-α, colony-stimulating factors, iNOS, IgG
  • Decreased histamine release from mast cells and basophils
  • Increased transcription of anti-inflammatory cytokines
  • Increased osteoclast activity

Main effects: COX-2 expression increased during inflammation and produces prostanoids. Decreased COX-2 expression and increased lipocortin/annexin production, which acts to inhibit PLA2.

174
Q

What is the effect of glucocorticoids on early and late inflammatory responses?

A

Glucocorticoids inhibit early inflammation response – redness, heat, swelling, pain, loss of function.

And inhibit late inflammatory response – wound healing and repair.

175
Q

List the clinical uses of glucocorticoids.

A
  • Treatment of inflammatory, immune or tumour related diseases
  • Emergency treatment of anaphylaxis, shock, asthma and trauma of CNS
  • Can be administered topically, orally, IV, IM or intralesional
  • Asthma
  • Recurrent airway obstruction
  • Eczema, rhinitis, allergic conjunctivitis
  • Hypersensitive states – severe allergic reactions to drugs or insect bites
  • Suppress graft rejection
  • Acute spinal injury
176
Q

What are the unwanted side effects of glucocorticoids?

A

Suppression of response to infection
Suppression of endogenous glucocorticoid production
Metabolic actions
Osteoporosis
Cushing’s syndrome

177
Q

How do glucocorticoids prevent T helper cell proliferation?

A
  1. Glucocorticoids suppress IL2 gene transcription
  2. Prevent Th cell proliferation
  3. Other drugs inhibit calcineurin activation to prevent IL2 production and Th proliferation, such as ciclosporin (aka cyclosporin) and tacrolimus.
  4. Ciclosporin can be given (sometimes together with glucocorticoids) for immune suppression in humans after organ transplantation.

Ciclosporin also used in vet practice, such as for atopic dermatitis in dogs but can cause kidney toxicity

178
Q

What are 5 examples of autoimmune diseases?

A

Addison’s disease
Systemic lupus erythematosus
Haemolytic anaemia
Myasthenia gravis
Arthritis

179
Q

What do T cells recognise?

A

T cells recognise small peptide fragments complexed with major histocompatibility complex molecules presented at the target cell surface.

180
Q

What do mature T cells express?

A

A T cell receptor together with either a CD4 and CD8 co-receptor.

181
Q

How do TCR, CD4 and CD8 recognise MHC?

A
  • TCR and co-receptors bind to different sites on the MHC molecules
  • A single MHC can be bound simultaneously by TCR and co-receptors
  • TCR is more peptide MHC recognition
  • CD4/CD8 enhances sensitivity
  • Unique systems has 1 ligand that the 2 receptors have 2 key interactions involved.
182
Q

What is the structure of the T cell receptor?

A
  • Alpha-Beta-T cell receptors are made up of 2 transmembrane chains – alpha and beta.
  • Each chain contains a variable and a constant region
  • TCRs bind to the peptide binding platform of MHC and mediate peptide MHC recognition.
183
Q

Describe the size of the T cell receptor repertoire.

A
  • Every individual possesses a large number of different TCRs.
  • Recent estimates suggest that every human has 25 million different TCRs.
  • Each TCR has its own unique antigen specificity.
  • This provides protection against a vast array of pathogens.
184
Q

What is clonal selection theory?

A

On recognition of antigen, individual TCRs undergo clonal expansion.

185
Q

What is crossreactivity?

A
  • Cross reactivity is effective immunity.
  • Cross reactivity caused pathogenesis of disease, such as the immunological basis of autoimmunity and transplant rejection.
  • Suggestion of cross reactivity being the underlying cause of autoimmunity has the mechanism ‘molecular mimicry’, microbial pathogens drive the expansion of self-reactive TCR clonotypes, causing autoimmune disease.
186
Q

What are the 3 signals delivered to naive T cells in T cell activation by APCs?

A
  1. Binding of the TCR and co-receptors, CD4 or CD8, to foreign peptide MHC.
  2. Co-stimulatory signal delivered by the same antigen presenting cell. CD28 on the T cell binds to B7 molecules on the APC.
  3. APC produces cytokines, which influence pathway of differentiation and produce different subsets of effector T cells that carry out different effector function, CD4 T cells in particular.
187
Q

What is immunological synapse?

A

Foci of interaction between T cells and antigen presenting cells. TCR-peptide MHC complexes concentrated here.

188
Q

Describe how engagement of TCR with specific peptide MHC triggers a cascade of signalling events that lead to T cell activation.

A
  1. The TCR is expressed in association with the CD3 complex.
  2. TCR-peptide MHC interaction results in phosphorylation of the immunoreceptor tyrosine based activation motifs in several CD3 chains.
  3. Phosphorylation of CD3 is mediated by protein kinase.
  4. Phosphorylated immunoreceptor tyrosine based activation motifs recruit and activate ZAP70, which triggers a cascade of signalling.
  5. T cell signalling events lead to the activation of transcription factors in the nucleus, which result in proliferation and T cell effector function.
189
Q

Describe T cell clonal proliferation and differentiation following T cell activation.

A
  1. T cells undergo a physical transformation from a small lymphocyte into a lymphoblast.
  2. Activated T cell starts to secrete cytokines and express cytokine receptors (IL-2 autocrine growth factor).
  3. The activated T cell starts to divide which generates lost of T cells with the same TCR.
  4. Most become effector cells but some also become memory cells, which are responsible for immunological memory.
190
Q

Describe the generation of T cell receptor diversity.

A
  • The T cell repertoire is generated by a process called somatic recombination.
  • This involves rearranging genes that encode parts of the complete alpha and beta chains.
  • The TCR-alpha locus: variable, joining and constant gene segments, V-alpha J-alpha C-alpha.
  • The TCR-beta locus: variable, diversity, joining and constant gene segments, V-beta D-beta J-beta C-beta.
191
Q

How are T cell receptors assembled?

A
  1. 1 variable, diversity and joining region gene is selected (TCR).
  2. The intervening intronic sequence is looped out and deleted.
  3. The C region is added by RNA splicing after transcription.
192
Q

Why does T cell receptor diversity arise?

A
  • Different variable regions combine with variety of D and J regions in the beta chain and J regions in the alpha chain. This is combinatorial diversity.
  • The way the segments join is not precise, this is called junctional diversity.
  • Nucleotides may be added when the joins in the DNA are made. This is called N-nucleotide addition.
  • Different alpha chains can combine with different beta chains.
193
Q

How do T cells develop?

A
  1. Migration form cortex to medulla of bone marrow.
  2. Differentiation to express a TCR with either CD4 or CD8.
  3. Positive selection of TCRs that can interact with peptide MHC complexes and others die by neglect.
  4. Apoptosis of TCRs that ineract with high affinity for slf peptide MHC in negative selection.
194
Q

How thymic maturation wasteful?

A

99% of immature T cells die by apoptosis and cell debris is phagocytosed.

195
Q

Define effector T cells.

A

Subsets of CD4 T cells are specialised to provide help or different pathogens.

196
Q

Describe TH1, TH2, TH17 and TFH effector T cells subsets.

A

TH1 – activate macrophages enabling destruction of intracellular organisms. Stimulate cytotoxic effect cells, CD8 T cells and natural killer cells.

TH2 – major role on providing B cell help to differentiate into plasma cells which secrete antibodies G, A and E.

TH17 – recruit neutrophils to sites of infection. Essential in response to fungi.

TFH – reside in lymphoid follicles. Important fr the formation and maintenance of germinal centres. Also in the regulation of B cell differentiation into plasma cells.

197
Q

What is immune deviation?

A

Immune responses can become polarised towards TH1 and TH2

198
Q

How does immune deviation occur?

A
  • TH1 and TH2 responses are mutually antagonistic.
  • TH1 cells produce IFNgamma which inhibits TH2 cells, leading to cytotoxic cell mediated immunity.
  • TH2 cells produce IL-4 & IL-13 which inhibit TH1 cells, leading to humoral immunity.
199
Q

What is a consequence of immune deviation?

A

Incorrect deviation is thought to contribute to pathology, such as a dog with leishmaniosis requires a strong TH1 polarization in order to control this protozoal infection. Failure leads to chronic multisystemic disease and death.

200
Q

How does the nature of the initiating antigen determine CD4 T cell differentiation?

A
  • All CD4 T-cells arise from a common naive CD4 T-cell precursor.
  • The way that antigen interacts with APCs (via PAMPs and PRRs) determines cytokine production by the antigen presenting cell, signal 3.
  • The variation in signal 3 causes naive CD4 T-cells to acquire different effector functions.
201
Q

What are CD8 cytotoxic T cells?

A

T-cells expressing CD8 differentiate into CD8 cytotoxic T-cells.

CD8 T-cells kill their target cells on recognition of specific peptide Major Histocompatibility Complex Class I.

Important for immunity against intracellular infection. Capable of killing cancerous cells.

202
Q

Describe the action of CD8 cytotoxic T cells.

A
  • CD8 T-cells have specialized lysosomes that contain cytotoxic proteins.
  • These are called perforin, granzymes and granulysin.
  • Perforin forms pores in the cell membrane causing cell lysis.
  • Granzymes enter cells and stimulate apoptosis.
203
Q

How do cytotoxic T cell form close adhesions to the target cell after recognition?

A
  1. Target and cytotoxic cells form membrane interdigitations and intercellular ‘tight junctions’.
  2. This forms a contained microenvironment in the narrow space between the cells.
204
Q

What happens when CD8 T cells specifically recognise a target cell?

A

The cytotoxic granules move and become polarised. Cytotoxic granules are released at the site of cell contact.

205
Q

What are the properties of memory T cells?

A
  • Are more sensitive to antigen
  • Express different cell surface markers
  • Are long-lived
206
Q

What is the role of natural killer cells?

A
  • Widely distributed at body surfaces
  • When viruses infect cells, they produce interferon-alpha (IFN-alpha)
  • This cytokine activates NK cells to kill the virus-infected cell
  • Killer cell immunoglobulin-like receptors (KIRs) are the dominant form of activatory and inhibitory receptors in humans
207
Q

How do natural killer cells kill virally infected cells or tumour cells?

A
  1. Downregulation or loss of MHCI at surface.
  2. So cell no longer engages the inhibitory receptor and no longer gives inhibitory signal to NK cell.
  3. In addition, disease cells also express ligands which engage the activating receptors on these NK cells.
  4. Activation of NK cells.
  5. Perforin and granzymes are released.
208
Q

Describe the properties and role of gamma-delta T cells.

A
  • Have a T cell receptor made up of gamma and delta chains
  • Develop in the thymus
  • Found at mucocutaneous surfaces of the body
  • In some animals and many in blood (cattle, sheep, pig)
  • Prominent in spleen (dog)
  • Location suggests they are important in ‘front line defence’ of the body
  • Particularly responsive to bacterial antigen such as Listeria, E. Coli, S. Typhimurium and Mycobacterium
209
Q

What is T cell therapy and the challenge of T cell therapy?

A

Aim is to harness the beneficial effects of T-cells and use them to treat diseases where there is an unmet need.

Challenges: we need to improve our basic understanding of the immune system to overcome the challenges of using cellular therapy.

210
Q

What are the advantages of T cell therapy?

A
  • T-cells have a potent ability to kill infected cells, especially useful in viral infections
  • T-cells have a potent ability to kill cancerous cells
  • T-cells can regulate the immune response, useful in autoimmunity and graft rejection
  • T-cell therapy may have a long lived effect, such as may be able to prevent cancer relapse
211
Q

What is adoptive T cell therapy? Outline its process.

A

A treatment used to help the immune system fight diseases, such as cancer and infections with certain viruses.

  1. T cells are collected from a patient and grown in the laboratory.
  2. This increases the number of T cells that are able to kill cancer cells or fight infections.
  3. These T cells are given back to the patient to help the immune system fight disease.
212
Q

Describe the process of genetically engineering T cells to enhance their activity.

A
  1. Isolate T-cells from blood & culture in vitro with cytokines.
  2. T-cells are genetically modified using vectors that encode receptors or molecules that enhance response to cancer cells.
  3. Most common vectors are gamma-retroviruses and lentiviruses engineered to be replication incompetent by the removal of genes that encode vital proteins.
  4. Genes of interest are incorporated in the genome of T-cells.
213
Q

What is the advantage of genetically engineering T cells?

A

It is possible to generate large numbers of cancer-specific CD8 T-cells in a very short space of time.

214
Q

What are the disadvantages of genetically engineering T cells?

A
  • Must identify a suitable MHCI restricted tumour rejection antigen
  • Must isolate a tumour specific TCR, which can be very challenging
  • This therapy is restricted by a specific MHCI so can only be used in a subset of patients
215
Q

How are T cells genetically engineered to express cancer specific chimeric antigen receptors?

A
  1. Operate in a non-major histocompatibility complex class I manner
  2. Extracellular domain is a single chain antibody
  3. Several different generations which vary in their cytoplasmic domains
216
Q

What are check point inhibitors and their applications?

A

T-cells within the immunosuppressive tumour microenvironment express a range of co-inhibitory receptors.

  • Engagement of these co-inhibitory receptors deliver a negative signal to the T-cells which prevents them from activating and destroying cancer cells.
  • Monoclonal antibodies that block PD-1 and CTLA-4 have been developed and are being used to treat humans with metastatic melanoma.
  • Amazing clinical responses have been seen in these patients although ~50% still do not respond so more work still required.
  • Other cancers are also being treated with these novel therapies e.g. lung cancers
217
Q

Distinguish B and T cell antigen recognition.

A
  • B and T cells are stimulated by different components of the same antigen.
  • T cells recognise processed peptide associated with MHC class I or II.
  • B cells recognise large, conformational determinants or epitopes, without the need to chop them up/no processed required.
  • May be free or non-specifically attached to the surface of an antigen presenting cell.
218
Q

What are the 2 B cell antigen recognition possibilities?

A

Thymus T cell independent – rare event

Thymus T cell dependent – ~99% of antigens are T cell dependent

219
Q

What follows activation of B cells by thymus independent antigens?

A

Results in B cell activation, which leads to maturation into plasma cells and antibody production, without requirement for T cell help.

220
Q

Why are T cells necessary to B cell activation?

A

T cell helps B cell become fully activated and there is no response to the antigen alone.

221
Q

What do naive B cells express?

A

In the periphery – spleen, lymph node and mucosa – naïve B cells express both IgM and IgD, and there are binding sites to the same antigen.

222
Q

What are the 3 signals required for activation of T cell dependent antigens?

A
  1. BCR binds to an epitope
  2. Co-stimulation: CD40L expressed by CD4 cells bind to CD40 on the B cell
  3. Cytokine produced by CD4 cells binds to cytokine receptors on the B cell
223
Q

What happens following B cell activation?

A
  • Transform into a lymphoblast
  • Ig class switch to IgG, IgA or IgE and no longer express IgM or IgD

Clonal proliferation and differentiation into plasma cells and memory cells. Plasma cells then secrete antibody that has the same specificity as the orginal smIgM or smIgD.

224
Q

Outline the steps of antibody production.

A
  1. Antigen presenting cells and T cells interact in the T cell areas of the secondary lymphoid tissues.
  2. Activated T cells move into the follicles and interact with B cells in the germinal centre.
  3. B cells that recognise antigens with high affinity, divide and mature into plasma cells, as long as they receive all 3 signals.
225
Q

What is B cell diversity?

A

The BCR like the TCR, is unique for each B cell and a vast repertoire of BCRs exist to account for all the potential antigen exposure.

226
Q

What does immunoglobulin class switching result in?

A
  1. Naïve B cells begin by expressing surface membrane IgG and surface membrane IgM.
  2. They interact with an antigen and activate, as they receive the 3 signals.
  3. They then undergo class switching.
  4. The activated B cell no longer expresses IgG or IgM.
  5. They now express a single class of immunoglobulin. 6. It will then differentiate into a plasma cell and subsequently start to secrete that immunoglobulin, which has the same specificity as the original antibody expressed by the naïve B cell, but is now just a different class of antibody.
227
Q

How is immunoglobulin class switching achieved?

A

Gene rearrangement

228
Q

Describe the process of gene rearrangement.

A
  1. Heavy chain genes
  2. DNA rearrangement loops out introns
  3. Transcription and alternative RNA splicing
  4. Translation (happens twice to make sure RNA with a mu on the end and some with δ on the end)
  5. Expression

IgM and IgD are expressed at the cell surface. However, the VDJ genes are the same, for example, antibodies have the same binding specificity.

229
Q

Describe class switching of mature B cells.

A
  1. 2nd DNA rearrangement is cytokine driven. Switches based on what would give the most effective immune response.
  2. mRNA transcription with RNA splicing
  3. Translation
  4. Expression as SmIgA
230
Q

Describe the affinity maturation of the B cell response.

A
  • As the antibody response matures, single nucleotide substitutions (point mutations) in Ig genes occur in somatic hypermutation.
  • Selection of B cells with high affinity BCRs from those with low affinity BCRs as the immune response matures and the antigen concentration decreases.
231
Q

What is immune suppression and its function?

A

Refers to switching off an immune response. Also called down regulation

Necessary to prevent unlimited and deleterious expansion of immune and inflammatory cascades. Prevents damage to normal tissue and protects our bodies.

232
Q

Name the 4 mechanisms of immune suppression.

A
  1. Antigen usage
  2. Antibody mediated suppression
  3. Hormonal regulation
  4. Suppressor T lymphocytes
233
Q

Describe immune suppression by antigen usage.

A
  1. Antigen
  2. Immune response
  3. Reduced antigen concentration
  4. Reduced immune stimulation
234
Q

Describe immune suppression by antibody mediated suppression.

A

a) Reducing antigen concentration – antibody combines with antigen, thereby reducing antigen concentration, which is linked to antigen usage/mechanism 1.
b) Immune complexes of antigen an antibody are immunosuppressive – antigens and antibodies from immune complexes, which are then removed by phagocytes that subsequently release suppressive substances, such as PGE2.

235
Q

Describe immune suppression by hormonal regulation.

A

The neuro-endocrine-immunological loop – effects that are mediated by soluble messenger proteins.

  • Stress > pituitary gland > adrenal cortex > cortisol > immune suppression
  • Cytokines > hypothalamus > temperature set point > fever
  • Some sex hormones can influence immune system.
236
Q

Describe immune suppression by suppressor T lymphocytes.

A

T lymphocytes can suppress immune responses caused by other T and B lymphocytes. Also called regulatory T cells.

237
Q

Outline an experiment to identify regulatory T lymphocytes.

A
  1. Inject mouse with high dose of foreign antigen
  2. 4 days later, remove spleen from mouse and make lymphocyte suspension
  3. Adoptive transfer to naïve recipient
  4. Immunogenic dose of antigen
  5. Makes weak serum Ab response to SRBC due to presence of suppressor T lymphocytes
238
Q

Describe the type of CD4+ and CD8+ T cells.

A

CD4+ T cells are the majority – spontaneously arising or induced types

CD8+ T cells less important – induced type

239
Q

How do CD4+ suppressor T cells work?

A
  • By regulating the balance of TH1 vs TH2.
  • Natural suppressors include: CD4+ and CD25+ T reg
  • Induced suppressors: CD4+IL-10 producing Tr1 cells
  • The TGF-beta producing induced suppressors
240
Q

Describe leishmaniosis as an example of immune deviation in infectious disease.

A

Leishmaniosis is not endemic in the UK but can see cases form animals that have travelled abroad, transmitted by sand flies in protozoal infection.

TH1 response is protective and TH2 response is not protective and results in severe disease.

241
Q

What are the natural suppressors and their function?

A

CD4+, CD25+, FOXP3+ Treg

  • Always normally present in the body and require direct contact to work.
  • Control of unwanted immune responses, such as autoimmunity
  • Can be identified by the fact that they express CD25+ and FOXP3+
  • Are found in humans and animals, such as cats, dogs and horses
242
Q

What are the induced suppressors and their role?

A

IL-10 producing Tr1 are induced suppressors – induced by an immune response and are called Tr1 cells.

Work via cytokines, not direct contact. Key cytokine is IL-10, which can be identified by their ability to produce IL-10.

TGF- producing TH3 is important in ‘oral tolerance’. Induced and work via cytokines, not direct contact

243
Q

Define hypersensitivity.

A

An individual who has been sensitized to a particular antigen by repeated exposure over time is susceptible to developing an excessive immune response (hypersensitivity reaction) on subsequent exposure to the antigen.

The same hypersensitivity mechanisms evolved for a useful purpose and are also utilised in appropriate immune responses to foreign antigen. They may be better considered as ‘immunopathological mechanisms’ rather than ‘hypersensitivity mechanisms’.

244
Q

What is the Gell and Coombs classification of hypersensitivity?

A

Type I = immediate

Type II = antibody dependent
- Cytotoxic type
- Anti-receptor type
- (Sometimes considered as Type V)

Type III = immune complex

Type IV cell-mediated or delayed type hypersensitivity (DTH)

245
Q

What is type I hypersensitivity?

A

Sensitisation repeated exposure to antigen or allergen over time.

  • Allergens may be derived from: grass pollen, house dust mites, animal dander, food, bee venom.
  • Allergic reactions generally occur when sensitisation to an allergen occurs  development of allergen-specific IgE
246
Q

Outline a type I hypersensitivity reaction.

A
  1. Dendritic cell captures allergen, which is processed and presented by MHCII.
  2. Dendritic cell migrates to local lymphoid tissue and triggers a T cell response (TH2).
  3. TH2 response provides B cell help (IL-4,9 and 13)
  4. Allergen specific N cells are activated > plasma cells that produce allergen specific IgE.
247
Q

Why is IgE an unusual antibody isotype?

A
  • Serum concentration of IgE is very low
  • Produced in small quantities
  • Short half-life (2 days)
  • Mostly found bound to the high affinity Fc-epsilon-RI on mast cells and basophils
  • Fc-epsilon-RI receptors are usually saturated with IgE despite low concentration of IgE in serum (high affinity)
248
Q

What are type II hypersensitivity reactions?

A

Subsequent exposure to the same allergen will cross link IgE which is bound to mast cells resulting in mast cell degranulation and a Type I hypersensitivity reaction.

  • Allergen cross-links IgE bound to FceRI
  • Intracellular signalling pathway leads to degranulation
249
Q

Describe the process of degranulation.

A
  • Only occurs upon cross-linking of bound IgE
  • Cytoplasmic granules fuse with plasma membrane > release of chemical mediators
  • Inflammatory reaction is induced by release of granule contents (histamine) and secretion of newly formed mediators (prostaglandins, leukotrienes, cytokines)
  • Immediate symptoms – bronchoconstriction, vasodilation, pruritis, 5-60 mins
  • Late phase symptoms – recruitment, eosinophils and macrophages, 4-24 hours
250
Q

Give examples of type I localised and systemic hypersensitivity disease.

A

Localised –
- Cutaneous (flea allergy, insect bites)
- Respiratory (asthma)
- Intestinal (food allergy)

Systemic
- Anaphylaxis (bee sting, penicillin reaction
- Severe and life threatening
- Bronchoconstriction, laryngeal oedema, vasodilation

251
Q

How is type I sensitivity regulated?

A
  1. Lack of Treg activity
  2. IgE production
  3. Eosinophil attraction
  4. Mast cell attraction
252
Q

What are the factors of type I hypersensitivity?

A
  • Age
  • Parasitism?
  • Genetic
  • Microbial infection
  • Stress
  • Allergen exposure
  • Immune dysregulation
  • Allergy often has a genetic basis, certain breeds are susceptible
  • Other influences are important as well e.g. age of exposure
253
Q

What is the hygiene hypothesis?

A

Suggestion that a lack of childhood exposure to pathogens and less childhood infections results in the increased incidence of allergy.

In individuals where you get naturally exposed to different infections, this induces TEG cells as they secrete cytokines and allows for bystander suppression. Cleaner environments will result in reduces expansion of Treg resulting in more allergic reaction.

254
Q

What are transfusion reactions?

A
  • Transfusing a DEA1- dog with DEA1+ blood, the dog will make an anti-DEA1 antibody response (induced alloantibody).
  • If a second transfusion of DEA1+ blood is given then the anti-DEA1 antibody will bind to the DEA1+ RBCs mediating cytotoxicity i.e. TYPE II Hypersensitivity.
  • Type B cats have naturally occurring anti-A antibody in their blood. If a type B cat is transfused with type A or AB blood this will result in a severe transfusion reaction.
255
Q

What is autoimmune haemolytic anaemia?

A

The immune system destroys the dogs RBCs via antibody mediate cytotoxicity.

256
Q

What is antibody mediated cytotoxicity?

A

Triggers by production of antibody that can bind to a target cell in the body.

257
Q

What is Grave’s disease?

A
  • Grave’s disease is hyperthyroidism in humans
  • TSH engages with TSH receptor causing to release thyroid hormones
  • In Grave’s disease, antibodies are developed that are specific to the receptors of TSH and cause stimulation of the receptor when it binds
  • Results in excessive hormone production/hyperthyroidism
258
Q

What is myasthenia gravis?

A

Disease of the neuromuscular junction of humans, dogs and cats.

  • Dog or cat have muscle weakness and excessive fatigue
  • Disease occurs when nerve ending interact with the muscle motor end plate
  • Normally, Ach produced
  • In disease, produce antibodies specific to Ach receptors and blocks Ach from the receptor
259
Q

How does antigen excess cause type III hypersensitivity?

A
  • Sensitisation leads to circulating antibody
  • Exposure to high concentration antigen (antigen excess)
  • Results in the formation of small soluble immune complexes which are found in the blood and circulate around the body resulting in systemic disease
260
Q

How does antibody excess cause type III hypersensitivity?

A
  • Sensitisation which leads to an excessive IgG response
  • After an individual has become sensitized then subsequent local exposure to allergen will result in:
  • Local immune complex formation (Ag and Ab)
  • Triggers local inflammatory response
261
Q

What is the Arthus reaction?

A
  • Excess IgG response
  • Antigen enters where excess is
  • Immunocomplexes
  • Inflammation, most commonly in the respiratory tract
262
Q

How does immune complex deposition result in complement activation and vasculitis?

A
  1. Immune complexes become deposited in the circulation
  2. Cause vasodilation and vasculitis and inflammation.
  3. Platelet aggregation
  4. Microthrombi
  5. Blood vessel occlusion
  6. Ischaemic necrosis
263
Q

Describe localisation of circulating immune complexes.

A
  • IC deposition occurs at certain predilection sites: small bore capillaries with a turbulent blood flow
  • Sites: renal glomeruli, uveal tract of the eye, skin at the junction between the epidermis and dermis, and synovium of the joints.
264
Q

What are the factors determining immune complex deposition?

A
  • Size of complex
  • If IC clearance mechanisms are overwhelmed
  • Nature of antigen
  • Nature of antibody
  • More likely to deposit at sites of high blood pressure & turbulent blood flow e.g. renal glomeruli
  • Damage to blood vessel walls (Endothelial lesions)
265
Q

What is feline immune complex glomerulonephritis?

A

Causes a protein losing nephropathy

266
Q

What is type IV hypersensitivity?

A
  • Involves mononuclear cells & cytokines
  • Delayed in onset 24 - 72 hours
  • Delayed type hypersensitivity (DTH)
  • Contact allergy (mainly in humans)
267
Q

How do type IV hypersensitivity reactions occur?

A
  1. APC take up antigen and present peptides, stimulating TH1 response
  2. IFN-y and requirement of other immune cells CD8 and NK (this is what stimulates a type IV reaction)
268
Q

Define tolerance.

A

Failure of the immune system to respond to a particular antigen.
Neonatal tolerance is the 1st demonstration of tolerance.

269
Q

Describe dizygotic cattle twins as an example of neonatal tolerance.

A
  • Share placental circulation but are genetically different
  • Each calf is exposed to each other’s alloantigens in utero
  • Become therefore tolerant to these alloantigens
  • Do not reject tissue grafts as a result
270
Q

Describe bovine viral diarrhoea virus as an example of tolerance.

A

Calves exposed to viral antigens in utero become tolerised to the virus, are born with the infection and become persistently infected.

271
Q

What is experimental neonatal tolerance?

A
  • Expose neonatal (new-born) mouse to foreign antigen by injection or transgenic expression
  • Are tolerant to the antigen
  • Immune system is tricked into thinking the antigen is self
  • Adult mouse would not mount an immune response to antigen
272
Q

What is adult tolerance?

A

Tolerance can also be induced in adults. Experimental induction of tolerance to foreign antigens in adult animals.

273
Q

Distinguish high and low zone adult tolerance.

A

High zone adult tolerance – a single high dose of antigen can induce tolerance in both T and B cells.

Low zone adult tolerance:
- Repeated injections of low dose antigen (dosage and route of admin is key)
- Induces T cell tolerance
- T cell more readily tolerised than B cells
- If antigen is T dependent, B cells are effectively tolerant anyway

274
Q

What is oral tolerance?

A

Failure to respond systematically to antigen previously given orally at specific dosage.

275
Q

How can oral tolerance be induced?

A

A foreign antigen is fed to a human or animal. When the same antigen is injected systematically then the individual will fail to respond.

276
Q

Describe an experiment that induces oral tolerance using 2 groups of puppies.

A
  • 1 group is fed ova – often used as an antigen to experimental approaches to tolerance
  • 1 group fed a different antigen, Derp1
  • Dogs fed ova prior to immunisation get no immune response
  • Dog fed Derp1 have an immune response
  • So oral tolerance is established
277
Q

How can oral tolerance be used as immunotherapy?

A
  • Systemic immune responses to an antigen are suppressed by feeding the antigen
  • Oral delivery of autoantigens or allergens may have therapeutic application
  • Could allow you to potentially treat autoimmunity?
278
Q

What is self tolerance?

A
  • Failure of the immune system to recognise self-antigens
  • T and B cells are educated to render them tolerant to self
  • Breakdown of self-tolerance results in autoimmunity
279
Q

Describe central deletion as a method of T cell self tolerance.

A
  1. Thymus selection of T cells to remove any autoreactive T cells from the population.
  2. This is not always successful and make it out to the periphery so important to have peripheral mechanisms for autoreactive T cells.
  3. These mechanisms are called peripheral tolerance.
  4. One of which is T regulatory cells, which are thought to prevent autoreactive T cells getting out of control and causing damage.
  5. There is also autoreactive T cell receive signal 1 and not signals 2 and 3, meaning it is unlike to activate and become anergic or die by apoptosis.
280
Q

What are the potential mechanisms for B cell self tolerance mechanisms?

A
  • Immature B cells encounter self-antigen in bone marrow?
  • Negative selection process?
  • Apoptosis of autoreactive clones?
  • Cannot activate without T cell help
281
Q

Define autoimmunity.

A

Failure of self-tolerance resulting in activation of autoreactive cells which may produce pathology and disease.

282
Q

Describe autoreactive T and B cells in a normal individual.

A
  • In normal individuals, you would expect to see autoreactive T and B cells being eliminated by peripheral mechanisms and not interact with self-cell and cause no autoimmunity.
  • We can take T cells and grow them in the lab against self-antigens.
283
Q

Describe autoreactive T and B cells in an autoimmune individual.

A
  • Can have a break down immunological tolerance
  • T regulatory cell are thought to play a role in tolerance so some defect in T regulatory cells causes lack of regulation and lead to activation to autoreactive T and B cells.
  • Attack of regular tissues and may have autoantibodies produced that also contribute to autoimmunity.
284
Q

What are the characteristics of autoimmune diseases?

A
  • Can involve almost all body systems
  • May be organ specific or multisystemic
  • Are highly important in humans
  • Occur in animals, particularly in dogs
285
Q

List some autoimmune diseases of companion animals.

A

IMHA (immune mediated haemolytic anaemia)
IMTP (immune mediated thrombocytopenia)
IMNP (immune mediated neutropenia)
Pemphigus
Polyarthritis
Myasthenia gravis
Thyroiditis
Diabetes mellitus
Systemic lupus

286
Q

What do immunopathogenic mechanisms of autoimmunity involve?

A

An immune response against auto-antigen:

  • Type II – cytotoxic or blocking antibodies
  • Type III – immune complex
  • Type IV – cell mediated
287
Q

What are the factors associated with autoimmunity?

A

Immunological abnormalities
Genetic background
Age
Gender
Environmental triggers

288
Q

How are immunological abnormalities a factor of autoimmunity?

A
  • Loss of self-tolerance – such as lack of regulatory cells.
  • Expression of MHCII and peptide by target cells – self tissue becomes a target
  • Epidermal expression
  • IFN-gamma - can be triggered by infection
289
Q

How is genetic background a factor of autoimmunity?

A
  • Some human autoimmune diseases ae more common in particular races and can run in families
  • Autoimmune disease occurs in particular breeds of dog
  • Which genes are associated with autoimmune disease?
  • The major histocompatibility complex – certain MHCs seem to predispose individuals to immune diseases.
  • Preferential presentation of self-peptide
  • Linkage to other disease genes?
290
Q

How is age a factor of autoimmune disease?

A
  • Autoimmunity occurs in middle ages to older dogs and cats
  • As age changes in a dog, lymphocyte function, CD4 T cells and B cells decrease and CD8 T cells increase.
  • Hypothesis is that some of the CD4 T cells that disappear are regulatory.
291
Q

Which environmental triggers factor in autoimmunity?

A

Season and climate
UV light
Air pollution
Diet and lifestyle
Stress
Drug administration
Infection

292
Q

What are some examples of drug therapy for autoimmune diseases?

A

TMS in Doberman and others – IMHA, IMTP and IMNP.

Felimazole in cats – IMHA, IMTP and ANA.

293
Q

What triggers autoimmunity?

A

Infection, concurrent or previous, triggers autoimmunity.

294
Q

What is an example of an experimental model of autoimmune disease?

A

Injecting a susceptible strain of infectious agent or peptide antigen from microbe is enough to start autoimmunity and can be injected into another mouse and also develop autoimmunity.

295
Q

How can an infectious agent trigger autoimmunity?

A

Superantigens – non-specifically stimulate autoreactive T cells and B cells.

296
Q

What is the innocent bystander effect?

A

Infectious agent attached to cell surface, such as RBCs in mycoplasma infection. Cell destroyed secondary to immune response to microbe.

297
Q

What is an immune complex disease?

A

A response to infection results in antibodies which then circulate around the body and accumulate in certain organs, such as renal glomeruli, triggering a type III hypersensitivity reaction.

298
Q

Explain induction of MHC and self-antigen expression.

A
  1. infection and tissue inflammation
  2. T cell cytokine, IFN-gamma
  3. MHC expression and self antigen
  4. T cell recognises and attacks
299
Q

What is molecular mimicry?

A
  1. Microbe and self-cell share antigenic component
  2. Presentation of microbial antigen activates self T cell
  3. Self B cell activated by microbial antigen
300
Q

What are the 2 basic principles of development of the immune system?

A

Immune system develops progressively in utero

The immune system can be activated in utero

301
Q

What happens in the first 100 days of pregnancy in the calf?

A

Blood lymphocytes can respond to mitogen in vitro in the 1st trimester, so:

  • Thymus develops
  • T cells in blood
  • Spleen
  • Blood SmIgM B cells
  • Complement
  • IFN-gamma/beta (antiviral)
302
Q

What happens by day 100 of pregnancy in the calf?

A

Can make antibody to anaplasma, Leptospira, parainfluenza 3, rotavirus, parvovirus by day 100.

303
Q

What happens from day 100-200 of pregnancy in the calf?

A
  • Can make serum IgG
  • Blood SmIgG B cells
  • Can make serum IgM
  • Tonsil
  • Peyer’s patches
304
Q

What happens by day 200 of pregnancy in the calf?

A

Can respond to campylobacter, chlamydia and E.coli beyond day 200. Normally born without serum Ig. Serum Ig at birth suggest in utero infection.

305
Q

What does passive transfer of immunity depend on?

A

Depends on type of placentation.

The classification of placenta according to how many cell layers separate the maternal and foetal circulation.

306
Q

Describe the horse and pig placentation.

A

Epitheliochorial. Has lots of layers and then intact epithelium and trophoblast layer

307
Q

Describe the ruminant placentation.

A

Synepitheliochorial. Has uterine connective tissue and epithelium and some fusion of trophoblast cells and epithelium

308
Q

Describe the dog and cat placentation.

A

Endotheliochorial. Has additional layer called maternal endothelium

309
Q

Describe the primate, rodent and rabbit placentation.

A

Hemochorial. Has trophoblast as the main layer

310
Q

How does primate, rodent and rabbit placentation impact passive transfer of immunity?

A
  • Born with serum IgG as adult level
  • Allows IgG to freely pass between maternal and foetal circulation and new-borns has a lot of IgG/similar to adult levels
311
Q

How does dog and cat placentation impact passive transfer of immunity?

A
  • Small amount IgG (5-10% adult level) passes
  • Maternal antibody ca interfere with the way dogs and cats respond to vaccination
312
Q

How does horse and pig placentation impact on passive transfer of immunity?

A
  • Contact of maternal endothelium, uterine connective tissue, uterine epithelium and trophoblast.
  • No Ig transfer possible due to robust layer
313
Q

How does ruminant placentation impact passive transfer of immunity?

A
  • Contact of maternal endothelium, uterine connective tissue, uterine epithelium and trophoblast.
  • No Ig transfer possible due to robust barrier
314
Q

What is the consequence of placentation of ruminant, horse and pig new borns?

A

Ruminants, horse and pigs have had no transfer of antibodies so are very vulnerable when born so very important for good colostral uptake.

315
Q

What are the species differences of colostrum?

A

Ruminant has lots of IgG and in non-ruminant has a dominating amount of IgA.

316
Q

How is colostrum absorbed and protected?

A

Gut permeability is increased from 6-24 hours.

Colostral protein protected by:
- Low proteolytic activity in the gut
- IgA secretory component
- Colostral trypsin inhibitors

317
Q

What are other things that may be absorbed from colostrum?

A

Complement, but when this happen sit may be non-functional. May also be some absorption of maternal lymphocytes but not proven.

318
Q

How are antibodies uptaken from colostrum?

A

Lymphocytes can express fc receptors, the binding of which causes the uptake of antibodies from the colostrum. Can cause peak serum in circulation of animal within 24 hours and can cause transient proteinuria by some loss of the proteins by the neonatal glomeruli.

319
Q

What are the species differences in absorption from colostrum?

A

Ideally want IgA to stay in gut lumen, as they protect gut mucosa.

In horse and pig, there is absorption of IgG and IgM and IgA stays within the gut mucosa.

But in the ruminant, there is absorption of IgG and IgM, and IgA is absorbed and re-excreted back into the gut lumen.

320
Q

What is the complication with neonatal vaccination?

A
  • Colostral Ig inhibits development of neonatal immune response until maternal Ig is catabolised.
  • Neonates cannot respond to vaccine until this has occurred
  • Giving too early affects the ability to respond
  • Repeat vaccination of neonates accounts for variability in colostral intake and catabolism.
321
Q

When are neonates vaccinated to avoid complications?

A

Vaccination at 8, 12 and 16 weeks to ensure that all are able to respond.

  • Vaccination too soon, internal antibody will interfere with the way the puppy or kitten will respond to vaccination.
  • Needs to be done before major susceptibility to certain infections.
  • This window varies between individuals and depends on the amount of colostrum absorbed.
322
Q

What are 2 possible disorder of the neonatal immune system?

A

Failure of passive/colostral transfer. Leads to enteric, respiratory disease or septicaemia.

Immunodeficiency

323
Q

What may a failure of passive/colostral transfer be due to?

A
  • Premature at birth and no colostrum
  • Premature lactation with no loss colostrum
  • Poor quality colostrum
  • Failure to suckle
  • Failure to absorb
  • Adequate colostral transfer determine serum Ig concentration after 24 hours of life

Treatment: administration of colostrum or plasma transfusion

324
Q

Describe the 2 types of immunodeficiencies of neonatal immune systems.

A

Primary: congenital, inherited and rare. 16 weeks of age

Secondary: acquired secondary to drugs, infectious, metabolic, neoplastic or inflammatory disease. Common.

325
Q

What are the clinical indications of immunodeficiency?

A
  • Chronic, recurrent infection in young, littermate animals
  • Infection at multiple sites
  • Failure to respond to standard antimicrobials
  • Infection with unusual agents
326
Q

What are possible immunodeficiencies in dogs?

A

30 immunodeficiency diseases (breed associated)

  • Weimaraner immunodeficiency syndrome – IgG deficiency
  • CLAD – canine leukocyte adhesion deficiency (Irish setters)

Congenital immunodeficiency in cats is rare.

327
Q

What is neonatal isoerythrolysis?

A
  • Isoimmune haemolytic anaemia
  • Can occur if the colostrum contains antibodies against the neonates erythrocytes
  • This can happen if dam has become sensitised to sire erythrocyte antigens or dam has spontaneously arising antibody that cross reacts with sire erythrocytes
328
Q

Explain an example of neonatal isoerythrolysis using a Q-A- mare and a Q+A+ stallion.

A
  • Given incompatible transfusion with Q+A+ blood (red blood cell antigens)
  • Sensitised to foal Q+A+ cells at first parturition
  • If mare is exposed to those antigens, it will cause an immune response and will produce antibodies against Q and A.
  • Could occur if mare has received a blood transfusion in the past containing Q and A (uncommon) or been expressed to red blood cells that express Q and A, which can happen after first parturition if the foal is Q and A positive.
  • Anti Q/A in colostrum
  • Haemolytic crisis in foal that is Q+A+
329
Q

Describe neonatal isoerythrolysis in cats.

A
  • Feline blood groups A, B and AB
  • All type B cats have high titred (antibody concentration) anti A
  • Type A cats sometimes have low titred anti-B
  • Neonatal isoerythrolysis can be caused in cats as a result:
  • A or AB kittens born to type B queens with colostral anti-A
  • Type B prevalent in Birman, Rex, BSH, Abyssinian, Persian and Somali
330
Q

Describe neonatal isoerythrolysis in dogs.

A

Rare in dog – but can be seen when sensitised DEA1- bitch mated to DEA1+ dog.

331
Q

What are the symptoms of neonatal isoerythrolysis in kittens?

A
  • Subclinical disease but may develop tail tip necrosis
  • 1-3 weeks post-partum
  • Severe haemolytic anaemia (Coombs positive)
  • Jaundice
  • Haemoglobinuria
  • Weakness, lethargy
  • Reluctance to suckle
  • Death within first few days of life
332
Q

How can neonatal isoerythrolysis be prevented and treated in kittens?

A
  • Prevent at risk mating by blood typing sire and dam
  • Determine level of anti-A in dam serum before birth by titrating against sire RBC
  • Prevent susceptible kittens access to colostrum for 24 hours after birth
  • Type A foster queen
  • Colostrum replacement with type A cat serum
333
Q

What are the problems with passive immunity?

A

Potential for hypersensitivity if given repeatedly.

Preformed antibody prevents recipient immune response.

334
Q

What is active immunisation?

A

The delivery of an antigen to induce an immune response and establish immunological memory.

335
Q

What are the properties of an ideal vaccine?

A
  • Cheap to produce
  • Stable
  • No side effects
  • Induce a protective immune response
  • Include range of epitopes to stimulate multiple clones of T and B cells
  • Induce immunological memory
336
Q

What are the properties of live vaccines?

A
  • Whole pathogen vaccines
  • Virulent - for example, orf in sheep
  • Attenuated
  • Heterologous
337
Q

Define attenuated.

A

So cannot cause disease. Done by heat, chemicals, alter growth conditions (medium, animal species), or (modern) genetic modification to attenuate the organism, for example, by modifying or deleting virulence genes.

338
Q

Define heterologous vaccines.

A

Antigenically related but adapted to another species (for example, measles – distemper, no longer used).

339
Q

What are the properties of killed/inactivated vaccines?

A

Whole pathogen vaccine.
Organism is antigenically intact but unable to replicate, due to formalin, alcohol or alkylating agents.

340
Q

Distinguish live and killed vaccines.

A

Live:
- Induce better immunity
- Fewer doses
- No adjuvant
- Greater risk of reversion to virulence or contamination
- Less stable

Killed:
- Multiple doses
- Must be administered with an adjuvant
- Stable
- Economical

341
Q

What are the properties of subunit vaccines?

A

Contain only specific metabolites or structural proteins from an organism that are known to cause an immune response.

342
Q

What are synthetic peptide vaccines?

A

Low immunogenicity (need adjuvant), monospecific immune response – although none currently available.

Type of subunit vaccine*

343
Q

How can subunit vaccines be made?

A

Subunit vaccines can be made using recombinant DNA technology. DNA encoding protein is inserted into a bacterial plasmid. Produce recombinant antigen protein.

344
Q

What are the properties of viral vectored vaccines?

A

Harmless viruses are used to deliver the genetic code of the antigens to cells of the body resulting in the production of protein antigens which stimulate an immune response. Viral vectors may be replicating or non-replicating.

345
Q

What are RNA vaccinations?

A
  • mRNA is injected into the body and taken up by cells where it is translated by the cell into antigens, which stimulates an immune response.
  • mRNA only lasts for a very short period of time (days) and is then broken down naturally and removed by the body.
346
Q

What are the advantages of RNA vaccination?

A

Can be produced much ,ore rapidly than conventional vaccines which is a major advantage in a pandemic situation.

Made using synthetic production methods, offering flexibility of pathogens are rapidly evolving.

347
Q

What are the disadvantages of RNA vaccinations?

A

Not very stable so need to be stored at a very low temperature, such as -70 degrees.

348
Q

What are DNA vaccinations?

A
  • Much more stable than mRNA vaccines
  • No licensed DNA vaccines currently
  • Often administered using a technique called electroporation to allow the DNA vaccine to be taken up by cells.
  • After DNA has entered cells, it is then translated into mRNA and subsequently protein antigens are made resulting in stimulation of an immune response.
349
Q

What is DIVA?

A

Differentiation between infection and vaccination

350
Q

How can vaccines and infections be distinguished from exposure titres?

A
  • Problem with FMD vaccination
  • Problem with FIV vaccine
  • (Fort dodge)
  • ospA Borrelia vaccine (recombitek lyme, merial)
  • IBR vaccine (infectious bovine rhinotracheitis) (gE negative mutant). Bayovac IBR, bayer
351
Q

How can vaccines be delivered?

A

Needle injection
Needle-free – transdermal delivery
Mucosal – intranasal
Aerosolization – poultry
Feed or water – poultry
In ovo – into eggs, poultry
Immersion – fish

352
Q

What are some guidelines when administering vaccinations?

A
  • Should be tailored to the individual animal or herd. Not every animal needs every vaccine.
  • Vaccines should be considered as core or non-core. All animals should receive core vaccines, but not every animal needs every non-core vaccine.
  • As many animals as possible within a population should be vaccinated with core vaccines in herd immunity.
  • Young animals require a series of priming vaccines followed by a 12 month booster.
353
Q

What are some vaccine associated adverse effects?

A
  • Transient post-vaccinal illness
  • Transient post-vaccinal immunosuppression
  • Local injection listen reactions
  • Hypersensitivity reactions
354
Q

What are the things to be aware of when administering vaccinations?

A
  • Not be given to pregnant animals unless licensed
  • Avoid giving vaccines to sick animals with weakened immune systems
  • Decisions on vaccination should be made in consultation with clients and considering data sheets and vaccination guidelines.
  • Always read and understand vaccine data sheet and guidelines.
  • Keep records of vaccination and report any adverse reactions to the company and/or VMD.
  • One dose fits all and animal size is irrelevant.
  • Vaccines rarely lead to adverse effects.
  • All licensed veterinary vaccines have proven quality, safety and efficacy.
355
Q

Why may there be a lack of efficacy in a vaccine?

A
  • Inappropriate administration
  • Administration to immunosuppressed animal
  • Batch of subnormal efficacy
  • Genetic non-responder
356
Q

What is the benefit of vaccination?

A
  • Control and possibly elimination of infectious disease
  • Bring disease under control and tackle pandemics
357
Q

What is intestinal rotavirus?

A
  • Viral pathogen in domestic species
  • Infects the epithelial cells of the gut
  • Will cause diarrhoea such as calf scours
358
Q

What barriers will a virus, such as intestinal rotavirus have to overcome?

A
  • Physical barrier – the epithelium
  • Intestinal secretions, such as mucus and bile salts
  • IgA and complement
  • Enzymes, such as lysozyme
359
Q

What happens to a viral infection, such as intestinal rotavirus, if physical barriers are overcome?

A

Innate immunity – non-specific defence, such as macrophages and natural killer cells.

360
Q

What happens if innate immune response is ineffective against a virus, such as intestinal rotavirus?

A
  1. Viruses often use receptors expressed by normal cells to gain entry and begin replication
  2. Infected cell expresses viral Ag on MHC class I. The virus particle is broken down into peptide fragments which presented at the cell surface by MHCI.
  3. Infected cell releases IFN-alpha/beta. This makes the uninfected cell produce anti-viral proteins for resistance.
  4. Dendritic cells takes up virus particles. Migrate to the local draining lymph node, such as mesenteric lymph. Then triggers an adaptive immune response.
361
Q

Describe the adaptive immune response to a virus, such as intestinal rotavirus.

A

For intracellular pathogens, need to stimulate a cytotoxic T cell response. In the lymph nodes:

  1. Antigen presenting cell, the dendritic cell has taken up viral antigens and has arrived at local lymph nodes.
  2. Dendritic cells can stimulate CD8 and CD4 T cells, as it can pick up MHCI and II.
  3. Dendritic cells also stimulating TH1 response which further stimulate CD8 T cells by producing IFN-gamma.
  4. B cells stimulated by T helper cells, producing antibodies.
  5. Once stimulated, immune cells need to home back to site of infection.
362
Q

Describe lymphocyte homing.

A

T and B cells that have clonally expanded in the mesenteric lymph nodes must get to the site of infection.

  • Achieved by the recirculation pathway
  • Activated cells go into the lymph and then blood.
  • Then find adhesion molecules on endothelial cells and home to site of infection.
  • Specific immune cells arrive at the site of infection.
363
Q

What barriers does a bacterial infection, such as E.coli, need to overcome?

A

To get through gut epithelium, E.coli must overcome physical barrier (bile, gut flora and gut motility for example) and cells of innate immune response.

364
Q

How can a bacterial infection, such as E.coli, colonise the gut?

A

E.coli bind to K88 and K99 pilus receptors to colonise the gut surface. Gamma-delta T cells provide protection to gut epithelium, particularly in ruminants.

365
Q

What effects can bacterial infections, such as E.coli, have on the body after colonising the gut?

A

Local toxins act on the intestinal tract causing a net loss of fluid into the tract, causing metabolic diarrhoea. Systemic toxins are endotoxins, which may cause serious symptoms.

Local inflammation caused by bacteria that invade the intestinal wall.

366
Q

What is needed against a bacterial infection, such as E.coli, if it manages to colonise the gut?

A

Adaptive immunity is required. APC transport Ag to Peyer’s Patch or mesenteric lymph nodes. Stimulate TH2 response, which provide help to stimulate B cells that can produce specific E.coli antibodies in the lymph nodes. The adaptive immune response must then home back to the site of infection.

367
Q

Describe the immune response against a parasite.

A
  1. Parasite antigens released and taken up by antigen presenting cells.
  2. Parasites are large but release antigens that can be picked up by dendritic cells.
  3. APC transport Ag to Peyer’s Patch or mesenteric lymph nodes to trigger an adaptive immune response.
368
Q

Describe the adaptive immune response against a parasite.

A
  1. APC present antigen and cause TH2 response, that stimulate B cells to produce specific antibodies.
  2. Activate parasite-specific immune response in PP/LN.
  3. Immune cells must home back to specific site of infection.
  4. Lymphocyte homing back to the site of infection.
  5. Plasma cells produce parasite specific IgG and IgE.
369
Q

Describe the action of parasite specific IgE.

A
  1. IgE binds to mast cells dje to mast cell high affinity for igE receptors. Causes:
  • Eosinophil infiltration
  • Oedema, degranulation, vasodilation, local inflammation
  • IgE sensitises mast cells
  1. Inflammation makes parasite detach
  2. Goblet cell produce mucous to coat parasites to make it more difficult for worm to persist.
370
Q

Describe the immune response if the parasite migrates on to tissues.

A
  1. IgG/E binds parasite and recruits phagocytes
  2. Extracellular degranulation of phagocytes damages parasites
  3. IgG neutralise parasite products
371
Q

What is leishmania?

A

An obligate intracellular protozoan pathogen which infects macrophages and replicates inside them. Infected macrophage or dendritic cells transport antigen to regional draining lymph nodes.

372
Q

Describe the immune response to a protozoan infection, such as leishmania.

A
  1. Infected macrophage transports antigen to regional LN.
  2. Activate leishmania specific immune response in lymph node.
  3. A TH1 response is required for an effective response against leishmania. TH1 release IFN-gamma, to activate ,macrophages to cause intracellular killing of leishmania.
  4. Plasma cells binds to antigens to mediate complement mediated lysis.
  5. These immune cells must home back to site of infection.