Cellular basis of immunity Flashcards

1
Q

Does everything living have an immune system?

A

yes, most basic have only phagocytes, then first lymphocytes appear as more advanced then lymphocytes split into T and B cells as well as phagocytes

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

Has adaptive immunity become more efficient in vertebrates?

A

yes

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

GALT =?

A

Gut associated lymphoid tissue

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

Thymus =?

A

Specialised primary lymphoid organ where T cells mature

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

Spleen does?

A

synthesises immunoglobulins and is reservoir continuing half the body monocytes

Makes dendritic cells and macrophages for the innate immune system

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

What is AID?

A

Deaminase mediating class switch recombination and somatic hypermutation of antibody encoding genes

Essential for removal of auto reactive B cells

Order from GALT have developed from most simple to most complex

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

Immune system number of cells, types and number of connections?

A

10^12

Less than 10

Infinite

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

Brain number of cells, types and number of connections?

A

10^11

2

10^14

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

Where do all immune cells originate from?

A

Hematopoietic stem cells in the bone marrow

Goes into the lymphoid lineage, melodic lineage or erythroid lineage

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

Serum = ?

A

Plasma without clotting factors

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

Blood = ?

A

55% plasma = water, dissolved proteins, glucose, clotting factors, electrolytes, hormones, CO2, and oxygen

1% WBC

44% RBC

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

Where is the immune system?

A

In the lymphatics system and lymph organs

White blood cells circulate with blood

Some WBCs and quite a bit of plasma leave the capillaries in tissues

Circulation of plasma and WBC through interstitial space/tissues and back via lymphatics to lymph nodes allows surveillance of tissues for forge in molecules and pathogens

Around 70% of the immune system is associated with the gut

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

Describe neutrophils eosinophils and basophils?

A

Attack a variety of pathogens by phagocytosis or with antimicrobial or with anti-microbials and histamine released after degranulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Describe plasma cells derived from B lymphocytes?

A

Secrete large amounts of antibody

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

Describe B and T lymphocytes?

A

Orchestrate the adaptive immune response and provide immunological memory

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

Describe monocytes and macrophages?

A

Phagocytes and regulate many aspects of innate and adaptive immunity

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

What does each pathogen have?

A

Antigens on its surface known as pathogen associated molecular patterns

Immune cells have invariant receptors on their surfaces known as pattern recognition receptors that recognise many of these PAMPS

Once a pathogen is recognised a cascade of events occurs activating an immune response to destroy it

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

Examples of PAMPs recognised by the innate system?

A

Cell wall components:
Peptidoglycan (bacteria)
Lipopolysaccharide (gram positive bacteria)
Galactomannans

Nucleic acid:
CpG motif DNA (bacteria)
ssRNA (viruses)
dsRNA (viruses)

Conserved surface proteins
Flagellin (bacteria)

Conserved stress proteins”
Heat shock proteins (bacteria)

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

Pattern recognition soluble receptors?

A
Natural antibodies
Complement
Pentraxins
Collectins
Ficolins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Pattern recognition cell receptors?

A
Toll-like-receptors
NOD-like receptors
RIG-like receptors
C-type lectin like receptors 
Scavenger receptors
N=formyl met-leu-phe receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Features of innate system?

A

Fast

Doesn’t require immunological memory

Activated by binding of PAMPS by PRRs on phagocytic cells

can discriminate self and non self

Receptors encoded in germline
Limited diversity

Typically recognise structures common to different microbes

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

Features of adaptive system?

A

Slower

Activated once the innate has been activated

Activated by a cascade of chemical signals from sites of infection , and physical interactions with phagocytic cells of the innate immune system known as dendritic cells that migrate from sites of infection into the lymphatic system to activate lymphocytes

Can discriminate self and non self

Receptors encoded by gene recombination - massive diversity

Typically recognise structures unique to different microbes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What decides the extent to how innate or adaptive system is used?

A

Severity, context and duration of disease, innate is mainly for something like a minor cut or splinter

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

What are cytokines and chemokines?

A

signalling molecules released at sites of infection by resident macrophages recruit neutrophils from the bloodstream leading to localised inflammation

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

Types of lymphocytes?

A

All key in adaptive immunity

B-lymphocyte/Bcell - express antibodies as B cell receptors

Helper T lymphocytes

Cytotoxic T lymphocyte

Natural killer T cell

T-lymphocytes express T cell receptors

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

What happens with B and T cells during an adaptive response?

A

Have receptors specific for particular pathogen molecules proliferate

Clonal selection and expansion forms the basis of protective immunity and immunological memory through which the body is protected against future attack

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

Homeostasis in which specific molecular interactions keeps the immune system in check?

A

Antigen receptors and antigens

Cytokines/chemokines and their receptors
Cell-cell interactions

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

we get infected or colonised in?

A
Cytoplasm
Intracellular vesicles of cells
Interstitial spaces
Blood or lymph
Epithelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

First barrier broken through during infection?

A

Skin

Cuts or burns

Main portals of entry is the mucosal epithelia of the gastrointestinal respiratory and urogenital tracts

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

Immunity requires?

A

The recognition and elimination/containment of infectious organisms

Most front line defences provided by the innate

Consists of molecules and cells that distinguish host cells from those of infectious agents by recognising conserved constituents of microorganisms

Activated within hours of contact and efficacy not significantly increased by previous exposure

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

Describe immune cell differentiation?

A

Cells of the innate and adaptive immune systems are derived from the pluripotent hematopoietic stem cell

The HSC divides to replace itself and produce a progenitor cell with the potential to give rise to all of the hematopoietic line ashes by generating further progenitor cells committed to progressively narrower ranges of differentiated fates

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

The erythroid lineage gives rise to?

A

erythrocytes and to megakaryocytic which shed fragments that form the platelets that initiate blood clotting

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

The myeloid lineage gives rise to?

A

Phagocytic and inflammatory cells of innate immunity

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

The lymphoid lineage gives rise to?

A

T and B cells of adaptive immunity and to natural killer (NK) cells which are specialised cytotoxic cells

Killer cells count as innate as don’t require activation

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

Hematopoietic stem cell lineages?

A

SCF = stem cell factor
TPO =Thrombopoietin (platelet production)
EPO = Erythropoietin (erythrocyte production)

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

What protects epithelial surfaces, especially the mucosa of the gastrointestinal, respiratory and urogenital tracts?

A

Basophils, mast cells and eosinophils

Mast cells have a sentinel role, whilst basophils and eosinophils are circulating cells recruited from the bloodstream

Whereas neutrophils destroy internalised micro organisms these cells typically provide immune defence against pathogens too large to be internalised

On activation they release the contents of their cytotoxic granules to the exterior creating an environment hostile to an invading organism or directly killing it

They also release histamines that mediate allergic and inflammatory responses such as coughing sneezing and vomiting

These expulsive responses may be mechanisms that are evolved to expel parasites

All 4 are known as the granulocytes

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

Who discovered phagocytosis?

A

Ilya Mechnikov (1898)

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

The phagocytic cells of the immune system comprise of?

A

neutrophils and macrophages which are effector cells, specialised to internalise and destroy micro organisms, and dendritic cells which internalise micro organisms for presentation to T lymphocytes of the adaptive immune system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Features of neutrophils?

A

Front line effector cells of innate immunity

After differentiation they circulate for a few hours before entering tissues to engulf microorganisms and destroy them via microbicidal products stored in vesicles

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

Features of macrophages?

A

Long lived cells that provide immune surveillance

They are derived from monocytes that circulate in the blood, differentiating as they leave the blood stream

Like neutrophils they ingest and destroy micro organisms

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

Features of dendritic cells?

A

Are involved in the induction of adaptive immunity

As immature cells they operate as phagocytes but rather than destroying the micro organisms they ingest their function is to display the ingested particles on their surface for recognition by T lymphocytes

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

Describe macrophage receptors?

A

Scavenger receptors - recognise particles released by dead or damaged tissues

Complement receptors and antibody receptors

Similar on dendritic cells

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

Activation of the innate immune system?

A

Directly through recognition of micro organisms or indirectly through recognition by cells of the adaptive immune system that then activate the innate immune defences

This is done as a lymphocyte of the adaptive immune system produces antibodies whose variable regions recognise a surface component of the bacterium A non variable region of the antibody is then recognised by a receptor of the phagocyte which in turn is activated engulf it. In this way, bacteria that have masked the conserved components can be recognised and destroyed by the innate immune system

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

Whats opsonin?

A

A general term for soluble components of the immune system that coat micro organisms and stimulate uptake by phagocytes

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

2 ways phagocytes are pivotal role in the immune system?

A

On activation by microbial surfaces they release cytokines and chemokines that amplify the response to infection

Cytokines and chemokines are signalling molecules. They increase the permeability of blood vessels and recruit additional cells and molecules of the immune system to sites of infection - the inflammatory response

Hence they activate the adaptive immune response

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

What does the chemokines IL8 (CXCL8) do?

A

From a macrophage

Recruits neutrophils, basophils and T cells to site of infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Recruitment of neutrophils to inflammatory sites?

A

Fluid matrices contain antibacterial peptides and toxic proteins that are constitutively secreted and further induced in response to cell wall components such as lipopolysaccharide

B-defensins in lung and skin
a - defensives in paneth cells of the intestinal tract
Lysozyme in fluids bathing the cornea

Cathelicidins (another group of antimicrobial defensins) are produced by activated neutrophils and epithelial cells in addition to antimicrobial activity they trigger signaling through the formyl peptide receptor an innate immune recognition element thereby aiding recruitment of neutrophils to inflammatory sites

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

what are fMLPs?

A

powerful chemoattractants and activators of neutrophils

Derived from BH2 terminal extensions of newly synthesised polypeptides

fMLP directs the transport of secreted proteins the possess NH2 terminal signal peptides

Cleaved by a signal peptidase following polypeptide transport and released into the extracellular space

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

what are fMLPs?

A

powerful chemoattractants and activators of neutrophils

Derived from BH2 terminal extensions of newly synthesised polypeptides

fMLP directs the transport of secreted proteins the possess NH2 terminal signal peptides

Cleaved by a signal peptidase following polypeptide transport and released into the extracellular space

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

What is chemotaxis?

A

neutrophils are attracted to bacterial chemical products like the peptide fMLP

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

Describe the inflammatory response?

A

Memory T cells monitor keep the tissues of the body under surveillance

If they detect no antigen they return to circulation via the lymphatic system

If there is an antigen it is processed by antigen presenting cell, and presented as MHC peptide complexes

This activates memory T cells

Which release cytokines such as TNF-a which act on local blood vessels

TNF-a activates the vascular endothelium

Loosens gaps in them so more blood leaks out, swelling occurs and looks more red due to RBC

The activated endothelium also releases adhesion molecules that allows phagocytes out the blood vessel

Takes 1-3 days

The phagocytes uptake the antigen and also can present it

Memory and effector T cells also leave the vessel

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

Describe cytokine signalling?

A

Cytokine receptor consist of 2 chains

Each having an extracellular binding domain

An a intracellular binding domain which has JAKs kinases on

Without cytokines these 2 receptors are not associated

Cytokines stabilise the heterodimer bringing them together, including the 2 JAK kinases

The JAK kinases can then phosphorylate the cytoplasmic tails

STAT molecules then bind to the chains at the bottom of the receptors which get phosphorylated by the JAKs

The STAT molecules can now dimerise and migrate into the nucleus where they can directly activate gene transcription

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

Describe rolling adhesion?

A

Blood vessel endothelium at sites of infection expresses selectin molecules (P-selectin and E-selectin) the selectins are triggered by cytokines released by sites of inflammation

Leukocytes such as neutrophils express the ligand for these selections in the form os s-Lex

These creates a weak bond with the selectins and the neutrophils rolls along the blood vessel forming and breaking bonds

These neutrophils will find somewhere they can squeeze for the endothelial cells out of the blood vessel

Only happens in veins

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

Overview of the innate system?

A

Immediate response minutes to hours

Semi specific, recognition through invariant specifc receptors (PAMP)

Includes Granulocytes DC/Mono/Mac
NK cells
TLR/NLR/RLH
Cytokines Chemokines Antimicrobial molecules& complement

No memory

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

Ovreview of the adaptive immune response?

A

Functional 4-7 days after exposure

Specific through epitope specific receptors (TCR, lg)

Includes T cells/B cells DC
TCR/BCR
Antibodies Cytokines Chemokines

Memory

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

Does the innate system help healing?

A

yes

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

Most abundant white blood cell?

A

Neutrophils followed by lymphocytes

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

What are the first responders to tissue detected microbes?

A

Neutrophils

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

What are the types of secreted molecules released by

A

Lysozyme - released in fluids of cornea, saliva and gut (Paneth cells)

Breaks down peptidoglycan, a component of the bacterial cell wall

Phospholipase A2 - breaks down phospholipids in cell membrane of bacteria

Antimicrobial peptides - Defensins - (alpha-defensins in Paneth cells of the intestinal tract and beta-defensins in lung and skin) – disturb the cell membrane of bacteria and fungi

Cathelicidins (another group of antimicrobial defensins) are produced by activated neutrophils and epithelial cells

Histatins (oral cavity, active against fungi)

Lecticidins (creates a pore in microbial cell membranes)

Complement: exists in fluids and blood:
capable of direct killing of pathogens through membrane attack complex (MAC)
also potentiates phagocytosis and the recruitment of other inflammatory mediators

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

More details on complement?

A

Small proteins that are synthesised by the liver, tissue macrophages, blood monocytes and epithelial cells of the genitourinary system and gastrointestinal tract

They circulate the blood as inactive precursors

Can diffuse into tissues

Capable of direct killing of pathogens through membrane attack complex

Potentiaites phagocytosis

Potentiates the recruitment of other inflammatory mediators

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

What’s the complement cascade

A

Is a part of the immune system that enhances (complements) the ability of antibodies and phagocytic cells to clear microbes and damaged cells from an organism, promote inflammation, and attack the pathogen’s cell membrane.

Its spontaneous and low level activation

If deposited onto pathogen = activation

If deposited into host cell = inhibited

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What does C3 convertase do?

A

Cleaves C5 into:

C5a: potent anaphylotoxin
C5b: initiates Membrane attack complex

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

What is the membrane attack complex?

A

or terminal complement complex (TCC) is a structure typically formed on the surface of pathogen cell membranes as a result of the activation of the host’s complement system, and as such is one of the effector proteins of the immune system.

In order to permeabilize both the bacterial outer and inner membrane and thus kill a bacterium,

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

Where does phagocytosis occur?

A

Phagocytosis occurs mostly in neutrophils and in monocytes/macrophages/dendritic cells

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

What is phagosome killing?

A

The phagolysosome is matured through sequential fusion with endosomes and lysosomes and becomes a very specialised intracellular compartment

The bug is digested and degraded

Unpleasant environment for microbes

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

How does phagocytosis occur?

A

Microbes are recognised by phagocytic surface receptors:

  1. PAMPS-PRR (Toll-likereceptors, C-type lectins, scavenger receptors)
  2. Opsonization
    Microbe coated with complement = binds CR3-CR4
    Microbe coated with antibody = antibody binds FcRecepors

Recognition and subsequent signalling trigger phagocytosis

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

Describe how the phagolysosome is matured from a phagosome

A

The phagolysosome is matured through sequential maturation with lysosomes and becomes a very specialized intracellular compartment with:

Low pH (pH=4.5): mediated by a V-ATPase proton channel

Accumulation of reactive oxygen species (NADPH oxidase - subunit gp91 or NOX2, and others)  oxidative stress

Accumulation of nitric species (iNOS)  nitrosative stress

Antimicrobial enzymes : cathelicidins, defensins, acid hydrolases, which are secreted into lumen of phagosome.

Low nutrients: no sugars, no micronutrients (Fe, Cu, Zn, etc), which are pumped out of the phagosome or sequestered (lactoferrin).

Phagosome maturation to phagolysosome takes 30-60 min.

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

What’s nutritional immunity?

A

When for example the phagasome pumps out iron which the microbe needs to live for nutrition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

How is the phagosome adapted in dendritic cells?

A

To perform antigen presentation, to degrade antigens and load them to bridge adaptive immunity

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

In neutrophils due to their specific granules?

A

The phagosome is basic (pH = 8 for about 30 min) to facilitate optimal activity of elastase and catches-in G

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

Extracellular killing?

A

Eosinophils (toxic granules):
They release mitochondrial DNA against bacteria

They also release granules are active against parasites, such as:
major basic protein (MBP)
eosinophil cationic protein (ECP)
eosinophil peroxidase (EPX)
eosinophil-derived neurotoxin (EDN)

Neutrophil extracellular traps (NETS) are also used catch microbes in sticky DNA fibrils

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

When killing a microbe is not possible what can happen?

A

Granuloma can contain microbes

Coordinated response of TH1 T-cells and macrophages which creates a layer of macrophages and outer layer of T cells which walls off the pathogen

Important in tuberculosis and in certain parasite infections

Means we can co live with the parasite

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

What organ is first to encounter blood after the gut?

A

The liver, gets rid of bad bacteria

This is done by liver macrophages called Kupffer cells using CRig functions as a pattern recognition receptor on Kupffer cells to directly bind gram positive bacteria via LTA, CRig-LTA is required for clearance of circulating bacteria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Example of regulation in the immune system?

A

Communication between all cells of the immune system
Phagocytosis occurs simultaneously with release of cytokines and chemokines
Complement deposition triggers phagocytosis and recruitment of other immune cells
Cytokines made by macrophages and dendritic cells trigger
recruitment of neutrophils and acute-phase response

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

Specific targeting and feedback mechanisms of the immune system?

A

Different PRR and cytokines  signal integration  adaptation of responses to each pathogen
Each microbe activates a set of PRRs different response based on ratio of signals allows a targeting of the response to pathogen and to tissue

Feedback mechanisms
Once other immune cells migrate into the tissue the cytokine and chemokine milieu modulates function of innate immune cells  differential activation of macrophages and monocytes ( and perhaps other cells)

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

How is host damage regulated/prevented?

A

If its chronic can cause a degenerative or auto immune disease

This is prevented by:
Host cells have mechanisms to prevent activation of innate immune killing

Intracellular signalling cascades triggered by PAMPS and DAMPS direct the magnitude and nature of the immune response in a tissue specific manner

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

What’s the danger theory?

A

posited that the primary function of the immune system is to sense and respond to danger in the form of signals (ATP, adenosine) derived from damaged tissue, regardless of the source of damage. Danger-Associated Molecular Patterns (DAMPS)

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

What is pattern recognition theory?

A

posited that lymphocyte activation is controlled by evolutionarily ancient system of germline-encoded pattern-recognition receptors (PRR) that detected conserved pathogen-associated molecular patterns (PAMPS) - predicted that PRR-PAMP interactions would induce costimulatory molecules on APCs.

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

When the immune system is de regulated?

A

: CYTOKINE STORMS, IMMUNE-MEDIATED DAMAGE, ALERGY, FIBROSIS

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

Overview of immune memory?

A

The adaptive immune system consist of T and B lymphocytes

The T lymphocytes and immunoglobulins, and antibodies that B-lymphocytes produce can recognise an unlimited number of different targets

Become effective only after a delay of 2 to 4 days on first encounter

Lymphocytes and lymphocyte products specific for a given organism then persist as immune memory and are rapidly protective on re-exposure to the same infectious agent

This property of the adaptive immune system forms the basis of the protective effects of vaccination

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

What does vaccination do?

A

The ability of the body to remember an infectious agent to
which it has been exposed is the basis for natural and artificial
immunity against a disease

Vaccination initiates a primary immune response, generating
memory cells without making the person ill

Later, if the same or very similar pathogens invade the body,
specific memory cells already exist

They recognise the agent and produce massive number of
lymphocytes and immunoglobulins that overwhelm the invaders

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

Specific functions of each cell in adaptive immunity?

A

Adaptive immune responses are mediated by lymphocytes of the lymphoid lineage which fall into two major classes:

T lymphocytes - kill virus-infected and cancerous cells (CD8+ T cells/cytotoxic T lymphocytes or CTLs

	or,

	       activate other cells of the immune system 		       (CD4+ helper T cells)

B lymphocytes - secrete immunoglobulins

Natural killer cells -capable of killing virus-infected or cancer cells immediately
without prior activation and without clonal expansion

They are generally regarded as part of the innate immune system
(because they don’t require prior activation)

Activation occurs via binding of activating receptors to surface molecules produced by cell damage e.g. cancer or that are encoded by infecting viruses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Definition of an antigen?

A

Any molecules or parts of molecules recognised by the variable antigen receptors of lymphocytes are known as antigens

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

Describe lymphoid lineage - cellular differentiation?

A

Mature lymphocytes that have not yet
encountered antigen are known as naïve
lymphocytes

Differentiation into effector cells is stimulated
by encounter with antigen and is preceded with
a vigorous proliferation that selectively expands
the numbers of those lymphocytes with receptors
specific for the inducing antigen

This is known as clonal selection of antigen-
specific lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Describe the lymphatic system?

A

The critical interactions between
activated dendritic cells and
lymphocytes occurs in lymphoid tissues

Lymphocytes enter lymphoid tissues 
from the bloodstream

Return to the bloodstream through 
the lymph if they are not activated

Migration through lymphoid tissues, 
and cellular interactions, are guided and
controlled by adhesive cell surface 
molecules whose expression is regulated 
by cytokines and chemokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

How do T lymphocytes recognise antigens?

A
T lymphocytes recognise antigens 
by means of T cell receptors 
generated during their differentiation -
they interact with APCs (antigen-
presenting cells - DC, macrophages and
B cells) via complementary ligands 

Each individual lymphocyte expresses
an antigen receptor of only one specificity,
but the total population of lymphocytes
in any given individual is collectively
capable of recognising virtually any antigen

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

What does CD stand for?

A

Clusters of differentiation

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

Features of the T cells CD4?

A

Marker of helper T cells

Is a co receptor for MHC call II molecules

These co-receptors play an important part in signal
tranduction on antigen binding by T cells

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

Features of CD8 T cells?

A

Marker of cytotoxic T cells

Co receptor for MHC call I molecules

These co-receptors play an important part in signal
tranduction on antigen binding by T cells

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

Features of dendritic cells?

A

Dendritic cells of the innate immune system play a crucial role in activating adaptive immunity

Begin life as phagocytes but when activated by conserved components of micro-organisms, or by inflammatory cytokines released by macrophages, become dedicated to displaying components of ingested micro-organisms for recognition by naïve T lymphocytes and subsequent differentiation of naïve T lymphocytes into effector T cells

Especially numerous in epithelia and at mucosal surfaces

Langerhans cells first recognised in the skin in the 19th century. They are a
subset of dendritic cells that reside in the keratinised epidermis for several months

Second population of skin-resident dendritic cells is found in the dermal layer - known as dermal dendritic cells or interstitial dendritic cells

In mucosal epithelium of the gut, dendritic cells are concentrated at specialised 
sites of antigen collection that overlie lymphoid tissues

Some have specialised surface properties that enable them to extend their long dendritic processes between the cells of the epithelium and into the lumen to sample antigens

These cells are directly exposed to ingested antigens and to commensal bacteria
as well as invading pathogens

These cells have a specialised role in preventing inflammatory responses to harmless gut residents
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

M cells do what?

A

deliver antigen from the lumen of the gut to the underlying tissue where dendritic cells cluster

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

Describe how the dendritic cells activates the adaptive immune system?

A

A dendritic cell displays a component of a
micro-organism for recognition by a T lymphocyte
bearing a receptor for that component. This
stimulates it to proliferate and differentiate into
an effector cell. When the effector cell recognises
the same antigen displayed on the surface of a
B lymphocyte, the T cell activates the B lymphocyte,
which in turn proliferates and secretes antibodies
that recognise the micro-organism

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

Describe T cell targeting?

A

Unlike B cells whose surface immunoglobulin molecules are adapted to serve
as antigen receptors, T cells are unable to interact directly with
intact antigen and so must interact with other cells of the immune system

T cells are focused on their target cells by cell surface proteins known as
MHC molecules because they are encoded in the major histocompatibility
complex (MHC)

MHC molecules carry fragments of antigen, usually peptide fragments of
proteins, from internal compartments of the target cell to the cell surface
and display them for recognition by the T cell

All naïve T cells are activated by antigen fragments displayed on MHC
molecules on the surface of dendritic cells, and after differentiation into
effector cells they are triggered to kill or activate cells displaying the same
complex of MHC and antigen

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

Whats the Major histocompatibility complex?

A

Chromosomal region containing genes that encode cell surface proteins
that regulate interactions among cells of the immune system

During transplant the more similar they are the longer it will take for the body to reject the new organ

There is class I molecules (Activate CD8 T cell) and class II molecules (Activate CD4 T cell)

These monitor different internal compartments of cells and are recognised bu distinct classes of T cells

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

2 internal compartments of cells that need to be monitored for pathogens?

A

The cytoplasm

All viruses and some important bacterial pathogens replicate
in the cytoplasm

Vesicles of the endosomal/lysosomal pathway

Contain internalised antigens derived from extracellular pathogens

In the specialised case of macrophages, vesicles may harbour bacteria
and some fungi (e.g. Cryptococcus) adapted to survive phagocytosis

The cytoplasm is monitored by MHC class I molecules

Vesicular compartments are monitored by MHC class II molecules

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

Features of MHC class I molecules?

A

MHC class I molecules are expressed on all nucleated cells
(not RBC) including APCs and bind peptides generated by cytoplasmic
proteases from cytoplasmic proteins

Their critical role is to display antigens derived from pathogens that
replicate in the cytoplasm, and antigen displayed on MHC class I
molecules is recognised by cytotoxic T cells, which kill the infected
cells

This system ensures the destruction of cells in which cytoplasmic
pathogens are replicating and is believed to be essential for the
elimination of some of these pathogens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What’s a cytotoxic T lymphocyte?

A

A specialised white blood cell responsible for eliminating unwanted body cells (eg. cancer cells) for example killing a cell infected with influenza virus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Describe recognition of antigen and MHC class I molecules by cytotoxic T cells?

A
Peptide fragments of pathogens, 
such as viruses, that replicate in the 
cytoplasm compartments of cells
are carried to the cell surface by
MHC class I molecules and are
recognised by cytotoxic T cells forming an immune-synapse, outer ring is an adhesion ring and the inner is a signalling zone and secretory zone

Upon activation, CD8 T cells produce:

  1. Perforins - proteins that form pores
    in the membrane of target cells
  2. Granzymes - proteases that induce
    programmed cell death on entry into
    target cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe MHC class II molecules?

A

MHC class II molecules are normally expressed only on cells of the
immune system, and in particular on professional APCs (B cells,
macrophages and dendritic cells)

They bind to peptides generated from internalised antigen in the
endosomal compartments of cells

Antigen derived from internalised micro-organisms by lysosomal
proteases and displayed by MHC class II molecules on dendritic
cells is recognised by naïve T helper cells at the initiation of immune
responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

MHC class II processing?

A

The 2 chains of it form in the endoplasmic reticulum

An invariant chains binds to the peptide grove formed by the 2 chains

Preventing unwanted peptides in the ER from binding

The invariant chains takes it through the ER and Golgi apparatus via a vesicle

Proteases are released inside the vesicle cleaving the invariant chain in 2 places leaving the CLIP peptide bound to the MHC class II molecule

Engulfed proteins or pathogens that are degaded in the vesicle can’t mind on to MHC class II as it’s blocked by CLIP

HLADM allows the CLIP to be removed, its present in the vesicle and acts as a catalyst

The pathogen peptides can now bind and the vesicle is taken to the cell surface to present as as an antigen

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

Naive helper T cells are first activated when?

A

They recognise antigen bound by MHC class II molecules on the surface of dendritic cells

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

Recognition of antigen and MHC class II molecules on macrophages by helper T cells?

A
Helper T cells recognise fragments
of a pathogen that has been internalised
by macrophages and have undergone
proteolytic digestion to generate
fragments that are carried to the
cell surface by MHC class II molecules.

The helper cell is then stimulated to
activate the macrophage to destroy
the internalised pathogen.

This is particularly important for defense
against bacteria that are able to grow
in the internal vesicles of macrophages

Helper T cell tells the macrophage via cytokines that what its doing is correct

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

Recognition of antigen and MHC class II molecules on B cells by helper T cells?

A
B cells internalise antigens bound
to their surface immunoglobulin
and deliver them to the endosomal
compartments of the cell where
they are digested into fragments.
The antigen fragments then bind
to MHC class II molecules which
carry them to the cell surface
where they are recognised by 
helper T cells, which then activate
the B cell to proliferate and 
differentiate into an antibody-
secreting plasma cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What is CD4 or CD8?

A

Coreceptor from Tcell receptor binding to MHC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

CD4+ T cell subsets?

A

There is TH1 and TH2 which release different cytokines which have different effects (picture in folder)

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

What do co-stimulator pairs do?

A

Enhance and sustain signal delivered by the MHC-antigen T cell receptor interaction (photo in folder)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Co-stimulator pair activation?

A

Professional antigen presenting cells present either MHC moelcules and co stimulator molecules such as CD80(B7.1)

When a complex is formed with the T cell signals are sent both ways, it tells the antigen presenting cell to produce more co stimulator pairs, and the T cell is induced to form pairs for the new co-stimulator pairs which can then form ligand making the connection stronger with the T cell

This then stimulates another co stimulator that is required when bound for full activation of the T cell

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

Summary of the immune response?

A

Pathogens manages to get through first layer of skin (through epidermis into dermis)

The bacteria establish an infection here

Phagocytic cells such as neutrophils and macrophages engulf the pathogen

Dendritic cells engulf as well and are stimulated by binding pathogens to leave the site of infection and migrate to a lmpyh vessel where the are drained into a lymph node

In the lymph node T cells are activated by the antigen presented on the dendritic cells and in turn activate B cells to release antibody

Effector T cells and antibody return to circulation

They return to the site of infection as inflammatory mediators have caused changes in the blood vessel endothelium

CD4 T cells activate macrophages to become more cytotoxic

antibody recruits complements of lysed bacteria directly and to opsonise them by inducing there uptake by phagocytes

In the case of a virus activated CD8 cells would kill any infected cells present

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

What does opsonise mean?

A

More susceptible to uptake by phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What are antibody molecules?

A

Host proteins produced in response to the presence
of foreign molecules in the body

Synthesised primarily by plasma cells (lymphoid lineage)
and are components of the adaptive immune system

Circulate throughout the blood and lymph where
they bind to foreign antigens

Antibody-antigen complexes are removed from
circulation primarily through phagocytosis by
macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Describe clonal selection of antigen specific lymphocytes?

A

The lymphocytes of any individual collectively express a large number of different receptors

Some cells bear receptors that recognise self-antigens. While still immature, they are eliminated

This process of clonal deletion is a critical mechanism for ensuring self-tolerance

Once mature, lymphocytes activated by recognition of foreign antigen differentiate into effector or memory cells through a process of clonal expansion

This is the mechanism for antigen-specific immune responses and immune memory

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

Receptor editing and clonal deletion of developing B cells?

A

The mechanism through which
self-tolerance is established differs between T cells and B cells

An immature B cell in the bone
marrow expresses membrane Ig antibody
together with Iga and Igb chains as the B cell receptor

If the BCR in is engaged by self antigens present in its local environment, maturation of the B cell is arrested and it stays in the bone marrow

Re-arrangement and recombination of genes encoding the BCR results in a new BCR possessing the original heavy chain but new light chain

This process is known as receptor editing

If the B cell is no longer auto-reactive, then it is exported to the periphery

If this BCR is still auto-reactive, then it can try again for a limited time before dying (clonal deletion)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Induction of B cell antibody response by T cells?

A

Germinal centres where B cells proliferate and undergo isotope switching hyper mutation

Germinal centres form from dendritic cells having antibody on their surface activating antigen specific CD4 T cells which proliferate into effector cells capable of activating antigen specific B cells

Once activated it proliferates to form a primary focus for antigen specific B cells

B cells from the primary focus migrate to nearby follicles whilst other B cells stay in the local area secreting antibody

B cells that enter to follicle proceed to proliferate rapidly under this time they also undergo somatic mutation to introduce new variation into the B cell receptor

The receptors are tested with their ability to bind to antigen those that fail to compete die

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Antibody structure

A

Large family of glycoproteins

Share key structural and functional features

Functional - able to bind both to antigens and to
specialised cells or proteins of the immune system

Structural - composed of one or more copies of a
characteristic unit that form a Y shape

So any molecules or parts of molecules recognised by the variable antigen receptors of lymphocytes are known as antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Whats the epitope (antigenic determinant)?

A

The specific region of the antigen bound by the variable region of an immunoglobulin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What’s the paratope?

A

The antigen binding region of an antibody

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What’s affinity?

A

Affinity is the measure of the strength of the binding of an
antigen by an antibody

Antibody binding to antigen is non-covalent and reversible

The affinity of an antibody (Ab) for an antigen (Ag) is related
to the ratio of the rates of the forward reaction for formation
of the complex to back reaction for decay of the complex

Like a reversible reaction in chemistry A level

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Papain cleavage on antibody?

A

Fab 1
Fab 11
Intact Fc 111

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

pepsin cleavage on antibody?

A

F(ab)2

Fc fragments

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

Antibody structure - the Y shape?

A

Heavy chain is top part of arm of Y
Light chain is lower part of arm of Y

Each Y contains four polypeptides

Two identical heavy chains
(55kDa each)

Two identical light chains
(22kDa each)

The four polypeptide
chains are held together
by disulphide bridges
and non-covalent bonds

The two domains that carry the antigen binding sites are known as Fab fragments (fragment having the antigen binding site) (the 2 arms) (antigen binds at the end of the arm)

The third domain is known as the Fc fragment (the fragment that crystallizes) (the base)

Hinge region in the middle of the arms aids movement

The ability to bind antigen
is determined by the N-terminal
domains. These differ considerably
between antibodies of differing
specificities and are therefore
termed ‘variable’ or ‘V’ domains - the end bits of each of the arms

Top part of the base of the Y is where complement binding happens

Bottom bit of the base of the Y is where binding to Fc receptors happens

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

What is hyper variability?

A
VH is top bit of the top arm of the Y, VL is the bottom bit of the top of the Y arm Sequence analysis of amino acids of
VH and VL regions reveals small
regions of hyper-variability within
four conserved framework regions
(FR1, FR2, FR3 and FR4)

In the 3-dimensional structure, the
hyper-variable regions form loops
that combine together to form the
principal antigen-binding surfaces

These surfaces are called the
complementarity determining regions
or CDRs (CDR1, CDR2 and CDR3)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Classes of immunoglobulin (different name for an antibody)

A

There are five main classes of antibody in serum

IgG, IgM, IgA, IgE and IgD

The five classes are divided on the basis of the
number of Y-like units and the type of heavy chain
polypeptide they contain (y, u, a, e and delta ) Greek symbols and associate same order as ones above

While there are five different types of heavy chains,
there are only two light chains (k or L) Greek again

One light chain always associates with one heavy
chain, so the number of light chains always equals
the number of heavy chains

There’s 2 heavy chains and 2 light chains per antibody

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

antibody in humans?

A

In humans, the IgG class has four sub-classes (G = gamma)

	IgG1, IgG2, IgG3 and IgG4

These have y1, y2, y3 and y4 heavy chains

The IgA class has two sub-classes

	IgA1 and IgA2

These have a1 and a2 chains respectively

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What’s a J chain?

A

Holds more than 1 antibody together

125
Q

What’s a secretory component?

A

In double antibodys

126
Q

Movement of antibodies across mucosal tissues with secretory IgA?

A

IgA is the most prevalent immunoglobulin in mucosal
secretions

Binds to antigens and prevents the uptake of bacteria and
toxins

Epithelial cells in the intestine mediate the transfer of
dimeric serum IgA to the intestinal lumen via the polymeric
Ig receptor

IgA dimer with J chain attached binds
to the polymeric Ig receptor, a transmembrane
protein expressed at the basal surface of
epithelial cells of the gut, airways and various
secretory glands

Binding of IgA induces transcytosis of the 
polymeric Ig receptor

Complex is delivered to the apical surface of
the epithelial cell and into the lumen

Proteases cleave the pIgR near the membrane,
releasing the majority of the extracellular
domain still bound to IgA dimer

The ‘secretory component’ protects IgA from proteases present in mucus and anchors IgA at the desired location

After opsonisation with IgA, the receptor Fca/mR mediates
the uptake of organisms by macrophages and dendritic cells

A second receptor FcaRI mediates uptake by neutrophils

IgA deficiency is a common immunodeficiency in humans

IgM can compensate for IgA deficiency by also binding to pIgR

IgM opsonised organisms are phagocytosed via Fca/uR

Activating Fc receptors signal through ITAMs

127
Q

What are ITAMS?

A

Antigen receptors, Fc receptors and some of the activating receptors of
natural killer cells signal via a common mechanism that is dependent on a conserved amino-acid sequence motif

This is known as the immunoreceptor tyrosine-based activation motif

The ITAM contains two precisely spaced tyrosines within a consensus sequence

When phosphorylated, the tyrosine residues provide a binding site for
one or two closely related intracellular tyrosine kinases

		Syk in most immune cells
		ZAP-70 in T cells

These have tandem SH2 (Src Homology 2) domains spaced at exactly the right distance apart to dock onto the two phosphotyrosines, and that activate signaling events downstream of the receptor

128
Q

Signal transduction by ITAM-containing immunorecetpors?

A

The surface immunoglobulin (Ig)
that serves as the B cell receptor for antigen
brings the receptor into proximity with a Src-
family tyrosine kinase, which is held in the
plasma membrane by a lipid tail and phosphorylates
ITAM tyrosines in the receptor Iga and IgB chains

This creates a binding site for Syk, which binds to
the doubly phosphorylated ITAM sequence

Once bound, Syk becomes activated by tyrosine
phosphorylation and it then phosphorylates
other signaling proteins that ultimately bring about
changes in the behaviour of the cell e.g. antibody
production

129
Q

Antibody-mediated cytotoxicity?

A

The two most widely recognised mechanisms of antibody-
mediated toxicity are:

  1. Antibody-dependent cell-mediated cytotoxicity (ADCC)

Antibody attracts cytotoxic cells by means of their Fc receptors

  1. Complement-mediated cytotoxicity (CMC)

Antibody binding results in the fixation of complement onto the target cell

130
Q

Opsonisation and phagocytosis process?

A

It requires triggering of specific receptors on the phagocyte

Fc receptors that bind antibody coated bacteria are one of the receptors capable of triggering phagocytosis engulfing it and forming a phagosome

Lysosomes move in to degrade the content in the phagosome

131
Q

What is complement?

A

The complement system, also known as complement, consists
of about 30 serum and membrane proteins that can mediate a
variety of immune reactions including:

Triggering inflammatory responses

Attraction of phagocytes to sites of inflammation

Degradation of membranes or virus envelopes

Stimulation of antibody production

132
Q

Activation of complement?

A

The active components of complement are generated from
inactive precursors by a cascade of proteolytic reactions

These are triggered through 3 different pathways:

The LECTIN pathway (recognition of carbohydrate
moieties e.g. by collectins such as mannose-binding
lectin)

  1. The CLASSICAL pathway (antibody binding to antigen in
    immune complexes)

The ALTERNATIVE pathway (directly at microbial cell
surfaces)

Activation of complement by any one of the pathways
initiates a cascade of cleavages in which each component
in the pathway is cleaved into two fragments:

The small fragment (a fragment)

The big fragment (b fragment)

The b fragment forms a subunit of the protease complex
mediating the next cleavage in the pathway

The reactions can be divided into early events, in which the
components are not the same for the same three pathways
and late events, in which they are identical

133
Q

Early and late events of complement formation?

A

The function of the early events is to generate two functionally
equivalent forms of a protease, known as C3 convertase

C3 convertase then initiates the late events to produce the
effector components of complement

C3 convertase covalently attaches to the cell surface at
which the complement activation was initiated

Cleavage of complement component C3 leads to effector
activation

In this way, the effects of complement activation are confined
to the infectious organism that triggered

134
Q

Pentraxins =?

A

Pentraxins = Pentameric serum
proteins that participate in innate
immunity and can also activate the
Classical pathway of complement

Antibody binds, C1r is activated by C1q C1r then cleaves and activates C1s

C1s cleaves C4 C4b attaches to cell surface C2 binds to C2 binds to C4b

C1s cleaves C2 bound to C4b producing C3 convert

C4bC2n and C3bBb conertases cleave C3 which provides C3a and C3b which are the effector actions, is an amplification loop for the alternative pathway as well

135
Q

What are collections and ficolins?

A

The epithelium of the respiratory tract is lubricated by a
layer of phospholipids and proteins known as surfactants

	Two of these, Surfactant Protein-A (SP-A) and Surfactant Protein-D (SP-D) are Collectins and function as opsonins,
	coating microorganisms (e.g. Pneumocystis in AIDs patients - Lecture 8) and stimulating uptake by phagocytes
The Collectin family also includes Mannose-Binding Lectin (MBL) - binds to mannose-containing carbohydrates on 
surface of viruses, bacteria and fungi

Ficolins bind to N-acetylglucosamine which are polysaccharides outside of microorganisms 

Each polypeptide chain of the collectin family of
proteins consists of an amino-terminal cysteine-rich region followed by a collagen-like region, an -helical neck region and a carboxy-terminal globular domain
(a C-type lectin known as the carbohydrate recognition domain [CRD])

The polypeptide chains trimerise

Both Mannose Binding Lectin and Surfuctant Protein-A are hexameric structures. The 
multiple arrays of CRDs give high affinity binding to
polysaccharide ligands in microbial cell walls

Particle recognition is translated into immune action:

1. 	By phagocytes, which are stimulated to internalise
	bound microorganisms or,
2.	By conformational changes in the collectin
	that activate pre-bound protease subunits and
	initiate the complement cascade
136
Q

Lectin pathway?

A

MASP = Mannose binding lectin associated serine protease

MASp-2 cleaves C4
C4b attaches to cell surface
C2 binds to C4b

MASP-2 cleaves C2 bound to C4b

Forming C3 convertase

C4bC2n and C3bBb conertases cleave C3 which provides C3a and C3b which are the effector actions, amplifies alternative pathway

137
Q

Alternative pathway?

A

C3b binds to microbial surface

B binds to C3b

D cleaves B bound to C3b forming C3 converts

C4bC2n and C3bBb conertases cleave C3 which provides C3a and C3b which are the effector actions

Amplifies alternative pathway

138
Q

C3a, C5a functions?

A

peptide mediators of inflammation, phagocyte recruitment

139
Q

C3b function?

A

Binds to complement receptors on phagocytes

Opsonization of pathogens

Removal of immune complexes

Can also results in terminal complement components C5b, C6, C7, C8, C9, which aid the membrane attack complex, lysis of certain pathogens and cells

C3b + C3 convertase → C5 convertase
C5 convertase + C5 → C5a + C5b
C5b + C6 + C7 + C8 + C9 ⇢ MAC

140
Q

What is hypersensitivity?

A

Undesirable reactions produced by the ‘normal’ immune response

Can be an allergy or auto-immunity

allergy = reacting to something not normally in the body

Auto-immunity - reacting to stuff in our body

141
Q

Different types of hypersensitivity reactions?

A
Type-I -
IgE
Causes degranulation of mast cells
Allergy
Asthma
Type II - 
IgG
Can recruit complement and attack things such as RBC
Autoimmune
haemolytic anemia
Type III - 
IgG
Binds to Fc receptor and recruits macrophages and neutrophils to the cell
Rheumatoid
arthritis
Type -IV
Doesn't use antibodies
Cellular effectors
Multiple
sclerosis 

Old system they are all mixed up in fact

142
Q

Allergy =?

A

• Excessive immune responses to harmless antigens (allergens)

	- Most allergic reactions are due to IgE-mediated responses

• ~40% of adults show an allergic response to least one environmental allergen:

Atopy: individuals that have a predisposition to become IgE-sensitized to allergens

No major common characteristics between allergens has been identified!

• Airborne allergens – small, soluble proteins (some with protease activity)

143
Q

Types of allergies?

A
Anaphylaxis
Acute urticaria
Allergic rhinitis
Asthma
Food allergy
144
Q

What’s asthma?

A

Airway hypersensitivity

Lung epithelium inflammation, smooth muscle contraction

145
Q

Cytokine environment orchestrated by what is crucial?

A

Which CD4+ Th subset it is

In allergy think its highly a Th2 mediated response can be termed Type 2 immunity

146
Q

Th2 response leads to?

A

IgE production that is specific to the allergen after CD4+ Th2 has stimulated a B cell

147
Q

Why can dendritic cells sample luminal antigens through the epithelial cells?

A

Can express tight junction proteins allowing interaction with epithelium

Epithelial cells can secrete factors that trigger DC activation:

Instructive cytokines

DAMPS - damage associated molecular pattern

148
Q

Effector mechanisms in IgE mediated allergy?

A

Mast cells: abundant in mucosal tissues (lung, gut)
and connective tissues (e.g. surrounding blood vessels)

Th2 tells B cell via Type 2 cytokines (IL - 4, IL - 5, IL - 13)to class switch IgE antibody, which forms plasma cells which take the antibody to the Fc receptors on mast cells that causes degranulation when stimulated by the allergen

Most IgE is cell bound (on FcεR) not in body fluids (unlike other antibodies)

149
Q

Mast cell granules?

A
Histamines
• Proteases
• Enzymes
• Cytokines 
• Prostaglandins
• Leukotrienes

In intestines causes fluid secretion and peristalsis that can case GI contents expulsion through diarrhoea or vomiting

Can decrease airway size increasing mucus

Increase blood flow and permeability - causing anaphylactic shock

150
Q

How are eosinophils different to mast cells?

A

Circulating rather than in tissue

Originate from bone marrow progenitors and are continually circulating
In health (% of circulating leukocytes):
Eosinophils ~ 2-6%
Basophils ~ 0.4% (very rare!)

Recruited to tissues via chemokines to sites of inflammation…

Activated by type-2 cytokines (especially IL-5), complement and antibodies

Release enzymes, toxic proteins, cytokines chemokines and leukotrienes

Characteristic feature of allergic disease (yet role is unclear)

151
Q

Time frame from these responses?

A

PEFR = peak expiratory flow rate = lung function

30 minutes huge dip in function due to mast cells but returns to normal in another 30 minutes

In late phase will dip not as steep around 8 hours

152
Q

Allergy can lead to airway tissue remodelling which is mediated by which events happening?

A

Too much mucus made:
Goblet cell hyperplasia

Airway smooth muscle:
Hyperplasia and hypertrophy

ECM deposition:
deposition of collagen

153
Q

Treating symptoms?

A

Antihistamines
Target H1 receptor

Adrenaline
Anaphylactic reactions, e.g. reforms endothelial tight junctions and relaxes smooth muscle.

Inhaled bronchodilators
Act on β-adrenergic receptors to relax constricted muscle

Immunosuppressive drugs (general anti-inflammatories) – corticosteroids
Suppress immune inflammation (can be given via inhalation)
154
Q

Developing therapie?

A

Antibody therapies:

Omalizumab (anti-IgE)
Reduces asthma symptoms;
• Allows for a reduction in inhaled corticosteroid (ICS) dose

Mepolizumab & Reslizumab (anti-IL-5 antibodies)
• Some evidence of efficacy, but only with patients where eosinophils are dominant (prednisone-dependent eosinophilic asthma).

Duplimab (anti-IL-4Rα – receptor for IL-4 and IL-13)
• Effective at controlling asthma with reduction in corticosteroid dose in mild to severe asthma patients that have high eosinophil blood count.

Low dose allergen therapies (allergen de-sensitization):
• Injection of low doses of allergen (that escalate).
• Mechanism unclear, though likely to involve Tregs and skewing toward IgG.

155
Q

Hygiene hypothesis?

A

Autoimmune disorders high in Western culture

Much cleaner

Could be due to microbiota

156
Q

The innate system recognises molecules that are?

A

Common to the pathogen is this lecture it is bacteria

Pathogen associated Molecular patterns (PAMPS)

The innate system is pre coded to recognise a limited repertoire of molecules

This enables it to respond quickly, but to provide a low level response

It kills by complement and interactions with phagocytes

157
Q

Overview of the 3 pathways of complement activation again?

A

In all pathways the first step is the recognition of microbial components:

Alternative pathway:
Recognised molecule - Spontaneous activation but various cell surface molecules play a role

Classical pathway:
Recognised molecule - Bound IgM or IgG, so relies on antibodies and is far more effective in individuals who have been previously exposed

Lectin pathway:
Recognised molecule - Carbohydrates such as mannose or N-acetyl glucosamine

158
Q

Early events of complement pathways results in?

A

C3 converts

C3 converts covalently attaches to the cell surface at which the complement activation was initiated

Turns into C3a (Inflammation - binding to receptors on mast cells, causing them to release histamine) and C3b (opsonization and phagocytosis)

In this way, the effects of complement activation are confined to the infectious organism

159
Q

After recognition - late events of complement pathways?

A

C3B forms part of the C5b convertase helping to later form the membrane attack complex which causes lysis of the microbe

C5b also produces C5a which causes inflammation

160
Q

Describe the membrane attack complex?

A

The Gram-positive cell wall tends to be thicker and more robust than the Gram negative cell wall

The MAC is much more active towards Gram-negative than gram positive bacteria

Ion influx and efflux causes the cell to lyse

161
Q

How do we know complement is needed to kill bacteria?

A

Can see that deficiency in being able to produce certain types of complement results in not being Protected against certain types of bacteria

162
Q

Indirect recognition of bacteria by phagocytes?

A

bacteria coated with antibodies are recognised by antibody receptors (Fc receptors)

Bactiera coated with C3b are recognised by C3b receptors

163
Q

Direct recognition of pathogen associated molecular patterns?

A

PAMPS are recognised Pattern recognition receptors such as Toll-like receptor

164
Q

Structure of a toll like receptor?

A

Extracellular domain - ligand recognition

Intracellular domain - signalling

(dimerisation is essential for signalling) so TL1/2 would be a heterodimer and TL5/5 would be homodimer

165
Q

Consequences of TLR recognition by phagocytes?

A

Activation of phagocytes which leads to:
Cytokine and chemokine production
Ingestion and killing

166
Q

TLR signalling and inflammatory responses?

A

Recognition of bacteria molecule by TLR

Dimerisation of the TLR

Signalling and inflammatory responses

167
Q

The adaptive system does what to bacteria?

A

Recognises antigens that are unique to an individual pathogen

It takes time to develop and mature but can provide a very powerful protective response

Normally recognises proteins or polysaccharides

168
Q

MHC presentational pathways?

A

Protein within the cytosol:
Processed by the proteasome generates peptides
Go to endoplasmic reticulum where become processed with MHC class 1 to form a complex which is recognises by CD8+ T cells

Activated CD8 T cels kill host cells infected with bacteria by releasing cytotoxins such as perforin, granzymes and granulysin

Protein taken up into the phagosome:
Endocytosis of extracellular protein in it's vesicle it forms a complex with with MHC class 11 from the ER, this complex is presented on the cell surface and recognised by CD4+ T cells

CD4+ T cells releases different cytokines that result in inflammation, macrophage activation, antibody response

169
Q

Bacterial diseases where CD8+ T cells play a role in protection because bacteria grow within host cells?

A

Tuberculosis
Salmonellosis
listeriosis
meliondosis

170
Q

What’s agglutination?

A

Clumps the bacteria so cleared better by phagocytes

Its triggered by antibodies

171
Q

What’s complement binding?

A

Antibodies bound to bacteria promote complement binding and complement activation by the classical pathway

Complement can now effect them

172
Q

What’s opsonisation?

A

Antibodies can make it easier for phagocytes to uptake bacteria

173
Q

Can antibodies neutralise toxins?

A

yes

174
Q

Overview of viruses?

A

Responsible for the commonest recurring infections of humans

Although usually transient, they have massive socio-economic consequences

Viruses that have ‘jumped’ species barriers (zoonotic) generally cause substantially greater mortality than endemic viruses that have adapted over greater evolutionary time to their hosts

Human Immunodeficiency virus from chimpanzees in Africa
Ebolaviruses from bats (via bushmeat e.g. duikers) in Africa
Severe Acute Respiratory Syndrome (SARS) SARS-CoV-1 coronavirus from horseshoe bats (via civets?) in China
Middle East Respiratory Syndrome (MERS) MERS-CoV coronavirus from camels in Middle East
Coronavirus disease 2019 (COVID-19) SARS-CoV-2 coronavirus from bats (via pangolins?) in China

175
Q

Epidemics are?

A

Epidemic: classification of a disease that appears as new cases in a given human population, during a given period, at
a rate that substantially exceeds what is ‘expected’, based on recent evidence

An epidemic may be restricted to one locale (an outbreak), more general (an epidemic) or even global (pandemic)

Pandemic: an epidemic that spreads through human populations across a large region e.g. a continent, or even worldwide

176
Q

Viral diversity?

A

Exist as obligate intracellular pathogens - dependent on host proteins
for replication

May contain as few as 3 to 4 to up to several hundred genes expressed
from single- or double-stranded RNA or DNA genomes

Genomes are enclosed within capsids (coat proteins)

Cytopathic (lytic) viruses e.g. poliovirus and influenza virus, lyse host
cells by inducing apoptosis or autophagy

Some cytopathic viruses e.g. herpes viruses can infect cells without
producing infectious virions until immunity wanes or until re-activated
by various physiological signals - known as latent viruses

Non-cytopathic viruses e.g. hepatitis B replicate without destruction of
cells

177
Q

GP encodes?

A

GP encodes Ebola virus glycoprotein (GP). Two forms:

  1. Trimeric trans-membrane complex targets virus to
    cells via receptor binding - allows virus to introduce
    its contents into monocytes/macrophages leading to
    inflammatory cytokine dysregulation
  2. Soluble dimeric form (sGP) secreted after infection –
    prevents neutrophil activation
178
Q

What’s a cytokine storm from ebola?

A

Immune cells get caught in endless loops releasing extreme levels of cytokines - proteins within cells which cause inflammation and attracting yet more immune cells

179
Q

What’s septic shock from ebola?

A

Infected cells detach from blood vessels causing massive haemorrhage. Loss of blood leads to kidney and liver failure

180
Q

Virus breaking and entering?

A

Like bacteria, viruses must overcome epithelial barriers
of skin and mucosa to establish infection

Intestinal viruses e.g. poliovirus enter via antigen-sampling
M cells

Respiratory viruses e.g. influenza virus and rhinoviruses
establish infection in the epithelial cells of the airways

Some viruses e.g. Zika, Dengue and West Nile viruses require
insect vectors for transmission

Bloodborne viruses e.g. HIV and hepatitis B can invade
through mucosa or epithelia following physical trauma

181
Q

Tissue specific receptors and viral tropism?

A

Viruses exploit specific molecules on cells as receptors for invasion

While some viruses bind widely expressed surface molecules, most viruses invade using receptors with restricted tissue expression patterns e.g. HIV

They are said to display TISSUE TROPISM

SARS-CoV-2 (COVID-19) accesses host cells via Spike Glycoprotein S binding to angiotensin-converting enzyme 2 (ACE2) on type II alveolar cells in lungs

182
Q

Virus immunity?

A

Major innate immunity is mediated via type 1 interferons*, complement and natural killer (NK)
cells

Major adaptive immunity is mediated via antibody
and cytotoxic T lymphocytes (CTLs)

Interferons are Group of proteins that induce host cell enzymes that affect transcription and translation of viral genes

183
Q

Adaptive immune response to acute virus infection?

A

Appearance of virus is followed
rapidly by a massive expansion of
virus-specific CD8 T cells

CD4 T cell expansion and contraction
also occurs but at a lower magnitude

Neutralising antibodies are produced
relatively early after acute infection
with cytolytic viruses

184
Q

What do neutralising antibodies do?

A

Prevent viruses leaving and entering cells

185
Q

Mechanisms of protective antibody responses to viruses?

A

The C1q/r/s complex couples
antibody binding to the
classical pathway of complement

Its structure is closely analogous to 
MBL and H-ficolin complexes
with MASPs (lectin  pathway)
186
Q

Features of aids?

A

Acquired Immune Deficiency Syndrome (AIDS) is caused by
the Human Immunodeficiency Virus (HIV)

HIV infects and destroys CD4+ T cells

GRID (later called AIDs) first recognised clinically in US in
1980s in intravenous drug users and gay men with no
apparent cause for impaired immunity but who showed
symptoms of Pneumocystis carinii pneumonia (PCP)

~50,000 people infected/yr (USA) and 39+ million deaths
globally since start of pandemic in 1980s

2/3 of infected people live in Sub-Saharan Africa, 1/5th in Asia

Vast majority of human AIDS infections caused by HIV-1

187
Q

Whats PCP

A

yeast like fungus pneumocystis jirovecii

Is opportunistic so found in healthy people lungs as well

Seen in people undergoing chemotherapy

188
Q

History of HIV?

A

HIV-1 originated in southern Cameroon from a simian immunodeficiency virus (SIVcpz), a retrovirus that infects non-human primates e.g. Pan troglodytes (chimpanzee) in West-Central Africa

SIVcpz introduced into humans through bushmeat activities with
subsequent mutation into HIV – requires one or more high-risk
transmission channels for human-to-human spread

Channels absent in Africa prior to 20th Century but growth of large Colonial African cities led to societal changes such as prostitution (with increased frequency of STDs (genital ulcer diseases e.g. syphilis)) that allowed sexually transmitted infection

Earliest documented case of HIV in a human in Congo in 1959

In US as early as 1966

189
Q

Important HIV glycoproteins?

A

gp 120 nicking glycoprotein

gp 41 transmembrane glycoprotein

190
Q

HIV epithelial entry?

A

HIV-1 can cross mucosal surfaces through M cells - prevalent in tonsils and rectal epithelia

It can also gain access via epithelia damaged by ulcerative infections, or by trauma or injection (i.v. drug use or blood transfusion)

Vaginal epithelia lack M cells - HIV gains access via interdigitating processes on
Langerhans cells

Epithelial cells in the small intestine express
CCR5, a chemokine receptor

HIV-1 binds to CCR5 (once tethered by
galactosylceramide) via the viral envelope
glycoprotein gp120 and the virus is transcytosed

191
Q

HIV transport to lymph nodes?

A

Once across mucosal epithelial barriers,
HIV-1 encounters sub-epithelial dendritic
cells

C-type lectin receptors (CLRs) on dendritic cells bind high-mannose N-linked
oligosaccharides on viral envelope
glycoprotein gp120

Interaction with CLRs initiates
dendritic cell migration to regional lymph
nodes

Once in lymph nodes, intact virus, internalised with CLRs, is re-exposed at the
surface and displayed to T cells

Regardless of the route of infection, the
predominant site of virus replication early
in disease is in the small intestine - this reflects the large number of activated T cells
that express CCR5 in that organ

192
Q

HIV getting into the CD4 T cell?

A

Entry of HIV into cells mediated by viral envelope glycoprotein gp120

Binds to CD4 and
chemokine receptors
(CCR5 or CXCR4) on
host surfaces

Binding with receptor
leads to a conformational
change in gp120 that exposes the transmembrane component of the envelope protein gp41

gp41 mediates fusion of
the virus envelope with the host cell membrane

193
Q

HIV infection cycle?

A

HIV infects cells via 2 cell surface molecules

CD4 is the primary receptor for the virus, whilst the chemokine receptors CCR5 or CXCR4 act as co receptors for the viral infection of macrophages and T cells

HIV binds initially to CD4 via the envelope like protein gp120, this interaction allows the uncoating of the virus and the entry of the nucleocapsid containing the viral genome

The viral reverse transcriptase copies the RNA genome of HIV into double stranded DNA which integrates into the chromosomal DNA of the host

In this state HIV is latent, it becomes transcripted when the host cell is activated this results in the accumulation of viral proteins as well as genome length transcripts of the virus

Viral proteins assemble at the cell membrane and bud off to form a new viral particle maturation occurs once its left the cell

194
Q

Immunological consequences of HIV infection?

A

Dendritic cells translocate HIV from mucosa within 30 minutes
of infection

A wave of viral proliferation in lymph nodes peaks 4-7 days after
infection

Viremia peaks at 14 days and all lymphoid tissues are infected
by 3 weeks

HIV is quiescent in resting CD4 T cells, but on T cell activation,
virus production is also activated and the host cell dies

> 99% of virus is produced by newly infected CD4 T cells

~107-108 CD4 T cells are producing virus at any given time

Loss of helper activities (mainly through cytokine stimulation)
means that maintenance of cytotoxic CD8 T cells is lost as
is the antibody response of B cells

Infection typically results in flu-like illness from days to weeks
after exposure - associated with a drop of CD4 T cells in the blood

The CD4-dependent antiviral cytotoxic CD8 T cell response develops
and is responsible for a prolonged period of stable viremia

This is known as the VIRAL SET POINT

Patients are normally asymptomatic until the CD4 T cell counts decline
further - infection of developing lymphocytes in the bone marrow and
thymus results in failure to replace lost T cells

Opportunistic infections begin when CD4 T cell counts reach levels
<200/l

Death then typically ensues in about 2 years

Common one is crypto coccus meningitis kills 600,000 lots of them with aids each year

195
Q

Overview of influenza virus?

A

There are 3 classes of influenza
viruses - A, B and C

Influenza A and B are major
causes of human disease

Only influenza A is responsible
for pandemic outbreaks

Influenza A viruses are avian
viruses that adapt to humans

The virus is spread by aerosol
droplets and establishes
cytolytic infections in the 
epithelial cells of the upper and
lower respiratory tract
196
Q

Features of influenza A?

A

Influenza A is a spherical or rod-
shaped enveloped virus covered
with 2 spike-like glycoproteins:

Trimeric hemagglutinin (HA)

Tetrameric neuraminidase (NA)

HA mediates binding to cell
surfaces and internalisation

NA cleaves sialic acid and
promotes viral release from
cells

197
Q

Features of influenza genotypes?

A

Influenza genotypes are diverse in birds (migratory waterfowl) and express a
variety of HA (H1 to H15) and NA (N1 to N9) subtypes

Avian viruses prefer receptors, primarily on intestinal cells, that have the -2,3
sialic acid linkage to galactose

Human viruses prefer the -2,6 linkage expressed on respiratory epithelia

Pigs express both types of linkage on respiratory epithelial cells

Influenza viruses enter the human population in regions where humans, pigs
and waterfowl, as wells as domestic chickens, are in close proximity

Pigs can be infected with both human and avian influenza, and exchange
of segments between viral genomes in the pig can give rise to variants expressing novel surface proteins together with human-adapted virulence determinants

198
Q

Immune response to infleunza virus infection?

A

Antibody mediated immunity:
Can bind to HA and block viral attachment
Can also bind to NA protein and prevent release of new virions

Cell mediated immunity:
CD8 T cells recognise the infected cell due to its MHC class 1 the T cell then causes cell lysis
199
Q

What’s antigenic shift and antigenic drift?

A

Epidemics occur when point mutations (red dots) accumulate in surface HA (ovals) or NA (diamonds)

This leads to ANTIGENIC DRIFT so
that, in many previously immune individuals, HA and NA are no longer recognised by neutralizing antibodies produced in response to earlier infections

Pandemics occur when avian influenza viruses and human influenza viruses infect pigs and re-assortment of the two genomes leads to the expression of avian HA or NA in a virus otherwise adapted to infect humans (ANTIGENIC
SHIFT)

200
Q

WHat’s hypercytokinemia?

A

A ‘cytokine storm’ (hypercytokinemia) is the systemic expression of a
healthy and vigorous immune system

Caused by release of >150 inflammatory mediators (pro-inflammatory cytokines,
anti-inflammatory cytokines, free radicals, coagulation factors etc.)

Precise mechanisms for cytokine storms are not known, but may be
caused by an exaggerated response when the immune system encounters
a new and highly pathogenic invader

They have the potential to do significant damage to body tissues and organs
e.g. if in the lungs, large concentrations of macrophages accumulate restricting
the airways

Cytokine storms occur in response to a number of infectious and non-infectious agents
e.g. avian influenza, adult respiratory distress syndrome (ARDS), sepsis and Ebola

201
Q

Overview of fungi?

A

Because they are eukaryotes, and share many similarities with mammals, antifungal drug design is difficult (cross reactivity and thus toxicity to the host).

Unicellular (yeasts_ and multicellular (filamentous) organisms

Ubiquitous components of terrestrial environments where they contribute to nutrient recycling

Endemic mycoses = living in geographic areas with distinct natural conditions

Many fungi dispersed by air borne spores that can reach terminal airspaces in the lung

Although many are transmitted from the environment, fungi that cause disease can also be normal commensal microbial flora = Candida albicans

Malassezia furfur and other species of this genus lives on skin = deregulated immune response associated with dandruff and may play a role in skin exacerbating skin immune pathologies

202
Q

mycosis = ?

A

Fungal infection of humans

203
Q

Top killers in immunosuppressed patients?

A

Developing countries are less affected by HIV-AIDS due to access to antiretroviral regimens
HOWEVER MORTALITY RATES IN SUSCEPTIBLE POPULATIONS ARE VERY HIGH
3% of patients develop infection
 dependent on transplant type, Candida most prevalent
30-40% mortality rate in transplant recipients in US

204
Q

Candida albicans virulence?

A

In response to microenvironmental signals of the host (changes in pH, cell density, exposure to serum and iron deprivation), the fungus switches from the yeast form to filamentous form.
The ability to switch morphologies has been linked to pathogenicity.
Both forms are likely to contribute to virulence:
Yeast form more easily disseminated through bloodstream
Filamentous form facilitates invasion and evasion of phagocytosis
Mutants unable to make the dimorphic switch are commonly non-pathogenic in animal models.

205
Q

Host recognition of fungal infection?

A

Innate recognition of fungi is mediated by interactions with conserved cell wall constituents (glucans, mannans, galactans).
PRR Dectin-1 (gene is CLEC7A) is highly expressed on the surface of dendritic cells and in lesser amounts on macrophages
It has a C-type lectin carbohydrate recognition domain that mediates
the recognition of beta 1,3- and 1,6-linked glucans on the surface of intact cells and zymosan (zymosan=cell wall fragments consisting of beta-glucans, mannans, mannoproteins and chitin)

So Dectin-1 is essential for immune system

Engagement of Dectin-1 results in tyrosine phosporylation of its cytoplasmic immunoreceptor tyrosine-based activation motif (ITAM) Syk (Il-10) and others (TNFalpha is Syk-independent)
Dectin-1 acts synergistically with TLRs to induce production of inflammatory cytokines and the production of reactive oxygen species (ROS)

206
Q

What are toll like receptors?

A

Dendritic cells and other immune cells must be able to distinguish between apoptotic particles generated by normal tissue turnover and particles that are indicative of infection

The molecules mostly responsible for making this pivotal distinction are those of the family of Toll-like receptors (TLRs)

Stimulation of immune cells through their TLRs leads to synthesis and secretion of proinflammatory cytokines, thereby initiating the inflammatory response that recruits both soluble immune components and immune cells from the blood

TLR stimulation of dendritic cells also induces the initiation of an adaptive immune response

Acts synergistically with TLRs

207
Q

Features of Mannose-binding lectin (MBL) and complement?

A

Candida expresses surface adhesins and mannoproteins that activate the alternative and lectin pathways of complement

MBL is a collectin, and is one of a number of serum proteins collectively known as opsonins

MASPS (Mannose-binding lectin-Associated Serine Proteases) couple collectins to the complement pathway

Before ligand binding, MASPs lack protease activity.

Binding of multiple carbohydrate recognition domains of MBL to the microbial cell surface leads to conformational changes in the molecule, which promotes proteolytic activation of the MASPs, which the initiate the complement cascade

208
Q

Defence against Candida albicans?

A

Activation of complement + Interactions of cell wall components, such as mannoproteins and beta- glucan, with phagocyte receptors such as TLR2 and dectin-1 stimulate
release of cytokines including IL-1, IL-6 and TNF.
phagocytosis by neutrophils and macrophages.

Phagocytosis triggers the production of appropriate innate fungal killing mechanisms

Production of cytokines by activated neutrophils, together with direct interaction of neutrophils with dendritic cells* within inflammatory sites, promotes the maturation of dendritic cells, = adaptive immunity is activated.

209
Q

Chromoblastomycosis and Fonsecaea pedrosoi?

A

non-fatal disease of skin and subcutaneous tissues high socio-economic burden
frequently occurs in tropical and sub-tropical regions of America, Asia and Africa
infection occurs following transcutaneous trauma (mostly farmers)
chronic disease that can take decades to develop (polymorphic skin lesion)
difficult to treat (antifungals, surgical debridement, with limited success)

210
Q

Cooperation between receptors on immune cells can result in?

A

Synergy and singla intergration

Resulting in enhanced cytokine production protective (curative anti-microbial immunity

211
Q

Why is it difficult to kill cryptococcal meningitis?

A

Cryptococcus is the exception in human fungal pathogens:

requires a Th1 protective response and killing seems to be more dependent on macrophages than neutrophils

independent of Dectin-1, since capsule hides glucan epitopes

no hyphae are produced in vivo. However giant cells are produced Titan cells.

Sugar capsule = invisibility cloak, harder for the body to make antibodies against sugar epitopes

212
Q

2 different types of eukaryotic parasites?

A
Proliferating unicellular = Protoza
Malaria
Trypanosomes
Leishmania
Toxoplasma 
Non-Proliferating multicellular helminths = Metazoa
Pinworm
Ascaris
Hookworm
Ascaris
Schistosomiasis
Filariasis
213
Q

Complicating factors of Parasites?

A
Numerous development stages
Numerous hosts
Vertebrate and Invertebrate
Many environments
Inside and outside host

Can then lead to long term chronic infections:

Immune evasion
Immune modulation
Immune pathology

= Absence of sterilising immunity

214
Q

Features of malaria?

A

Spread mainly by female Anopheles mosquitoes
Obligate intracellular parasite
All of the symptoms of severe malaria occur during the erythrocytic stage of infection
The liver stage is clinically silent
Different immune responses are required against liver and blood stages of infection
Stronger immune responses develop to the blood stage of infection
Precise mechanism of how malaria infection causes sudden and life-threatening complications is unclear

Cerebral malaria is when it gets into the brain 15-20% case fatality rate

215
Q

Features of Leishmania?

A

Primary host is the sandfly

Numerous different species of Leishmania parasites, each of which can give rise to a different type of disease

Obligate intracellular parasite that primarily invades macrophages

Can result in Mucocutaneous Cutaneous

216
Q

Features of Trypanosomes?

A

Extracellular parasite

• Causes “sleeping sickness”

217
Q

What does some parasites being intracellular and some being extracellular mean?

A

Too different for adaptive immune system to work on different ones as well

But there are still similar themes to protective immunity

218
Q

Innate cell killing of parasites?

A

Neutrophil mediating killing:

Neutrophil extracellular traps…
(Trojan horse)

Macrophage:

PRR activation and take up parasites (and infected neutrophils) via phagocytosis and enter phagosome

Killing:
Reactive oxygen species (ROS), triggered by phagocytosis
Production of nitric oxide…
Induced by IFNγ (Need help from Th1 CD4+ T cells!)

219
Q

How do parasites live inside macrophages?

A

Subversion and evasion

Sand fly salivary proteins protect against neutrophil mediated death

220
Q

Against parasites what type of T cell is better at killing them?

A

Th1 cell rather than a Th2 cell

The Th1 produces IFN-y cytokines that activate M1 macrophages which produce iNOS which produces nitric oxide which kills the parasites

Regulatory T cells can also suppress immunity and macrophage function

The effector functions of the pathogen specific CD4+ T cells determines the outcome of disease

221
Q

Immune response to plasmodium parasites?

A

Skin and liver stage - clinically silent and no hint of acquired immunity.
Symptoms appear during blood stage
Rupture of iRBCs and the release of merozoites = clinical symptoms of malaria
Variant surface antigens (VSA) – binding to endothelium and rossetting of RBC..
Immune responses:
Th1 CD4+ T cells, NK cell secrete pro-inflammatory cytokine
macrophages production of NO
B cell producing antibody (blocks invasion)

But VSA antigen shift – immune evasion…

222
Q

Trypanosome evasion via antigenic variation?

A

Trypanosome population can express up to 100 VSGs at any time.

And this can be shed (avoiding complement)

Sophisticated immune evasion strategy…

223
Q

3 types of metazoan parasites?

A

CESTODE - Tapeworms, Taenia, Echinochoccus

NEMATODE - Round worms, Filaria, Ascaris, Hookworm,Trichinella

TREMATODE - Flat worms, Schistosomiasis

Gastrointestinal, vs. tissue dwelling, vs. both!

224
Q

TH2 T cells needed to kill worms instead of TH1, explain?

A

Key events causing response:

Parasite damage

  • Tuft cells in intestine
  • Epithelial cell response
  • Parasite antigen stimulation on APCs

Impact of type-2 cytokine:

Stimulates mast cell degranulation (increase gut motility, fluid secretion)

Increase number of goblet cells –mucus hypersecretion (trap parasites).

Release of toxic proteins, directly harm parasites.

Increased epithelial cell turnover and intestine peristalsis.

All lead to parasite expulsion

Type 2 causes collagen synthesis and fibrosis due to arginase converting argine

225
Q

Granuloma responses?

A

Caused by macrophages

226
Q

Can parasite products be used as treatment?

A

yes - they can suppress the immune system in useful ways

227
Q

Overall summary on parasites?

A

Type 1 immunity for protoza

Type 2 immunity for Metazoan

Balanced immunity is essential to avoid host damage

228
Q

Is there resistance against vaccines?

A

Low levels of resistance to vaccines are a consequence of the timing of the treatment (prevent rather than treat infection) and the multiplicity of target sites

229
Q

Main types of vaccine?

A

Live vaccines
Living, but cannot cause disease

Killed vaccines
Killed by heating, or exposure to chemicals

Sub-unit vaccines
Fragments of the microorganism e.g. proteins (or polysaccharides)
Almost always antibody mediated
In some cases an immune response against a single protein antigen elicits protection

Nucleic acid vaccines (DNA or mRNA)
Direct protein expression after injection

Vectored vaccines
DNA encoding the sub-unit is cloned into another “safe” microorganism

230
Q

What is formaldehyde detoxification?

A

Removes toxicity of a protein

The immunogenicity of the protein is preserved

Problems:
Must balance cross linking with retention of antigenic structure

Batch to batch variation

Reversion

Requires pure toxin at the starting point:
Needed to grow pathogen
Need to purify and handle the toxin

231
Q

What are genetic toxoids?

A

Modern approach to subunit vaccines against toxins

Modify the gene so that it encodes a non toxic (but immunogenic) protein

232
Q

Describe e toxin?

A

causes enterotoxaemia in livestock

Produced in intestine crosses gut wall, targets kidneys and brain causing death

Can mutate the toxin now 1000 fold less toxic

Is a vaccine

233
Q

Diphtheria toxin CRM197 genetic toxoid?

A

Mutation in the catalytic A-subunit blocks activity

Glycine to glutamic acid substitution at residue 52

234
Q

Advantage of genetic toxoids?

A

Produce in harmless bacterium

High yields

Molecular structure more similar to active toxin

Reproducible properties

No reversion

235
Q

Sub unit vaccine against plague?

A

F1 antigen - capsule and V-antigen part of the type 111 system are the protective components

Then produce F1 and V antigens using genetic engineering

Together produces solid protection

236
Q

covid vaccine?

A

The spike S1 protein induces protective immune responses

Some antibodies against S1 protein epitopes are protective others make the disease worse

237
Q

What’s reverse vaccinology?

A

Predict vaccines from the genome sequence

Neisseria meningitidis causes globally 1.2 million
invasive disease cases and 135,000 deaths per year
• Serogroup B strains are the main cause of disease in Europe, New Zealand, Australia, Argentina, Canada, Japan
• Traditional vaccinology had failed to identify a vaccine

pSORT predicts cellular location so can help us find proteins that will be on the surface will give lots of proteins then you have to test them

238
Q

Meningitis B vaccine?

A

H binding protein (fHbp), fused with NA2091 protein, binds human factor H, a negative regulator of the alternative pathway of complement activation.
NadA, major adhesion protein involved in colonization, invasion, and induction of pro-inflammatory cytokines.
NHBA, heparin-binding protein that increases resistance against the bactericidal activity of human serum
PorA 1.4 Porin, A major outer membrane protein and the target for bactericidal antibodies

239
Q

Sub unit vaccines are effective at stimulating protective antibodies which will?

A

Agglutination

Opsonization

Neutralization

Activation of complement

Inflammation

Antibody - dependent cell mediated cytotoxicity

240
Q

Is there immunological cross reactivity between the different polysaccharides on bacteria?

A

No they are unique

241
Q

Examples of polysaccharide vaccines?

A

Streptococcus pneumoniae - pneumonia
• Haemophilus influenzae - pneumonia
• Neisseria meningitidis - meningitis
• Salmonella typhi – typhoid fever

242
Q

Features of Streptococcus pneumoniae?

A

Major cause worldwide of community acquired pneumonia, bacterial meningitis, bacteraemia and otitis media

Causes disease in young and elderly

Antibody against the capsular polysaccharide is protective

Antibody causes:
Opsonising Agglutinating Complement fixing

243
Q

3 problems with S.pneumoniae vaccines?

A

Polysaccharide diversity so doesn’t work on slightly different strains

  • At risk populations respond poorly to vaccines
  • Poor long-term memory
244
Q

How do they get around polysaccharide diversity?

A

Pick the serotypes most likely to cause disease and isolate these polysaccharides

Created a 23-valent vaccine

245
Q

Are polysaccharides T cell independent antigens?

A

Yes

No involvement of antigen presenting cells, CD4+ or CD8+ T cells
• Polysaccharides interact directly with B-cells
• Clonal proliferation of B-cells
• Production of IgM antibody
• Poor antibody responses in children & in the elderly
• no memory

Can fix this by chemically coupling it to a protein which means it can now be processed

246
Q

Mechanisms of T-cell activation by glycoconjugate (adding the protein) vaccines?

A

Glycoconjugate is internalized into an endosome of an antigen presenting cell (e.g. B cell).
• processedintoglycanP-peptides.The polysaccharide is broken down into ~10kDa glycans
• MHCII presentation of glycanP-peptide by MHCII to the αβ receptor of CD4+ T cells (αβTCR).
•The T-cells involved are called T cells carb
•Activation of the T cell results cytokines which mature the B cell to become a memory B cell (especially IL2 and IL4)

247
Q

Protein used to bind to the carbohydrate?

A

diphtheria toxin CRM197 genetic toxoid

• glycine to glutamic acid substitution at residue 52 abolishes toxicity

248
Q

Diseases where CD+ T cells play a role in protection?

A

Diseases where the microbe grows within host cells

tuberculosis
• salmonellosis • melioidosis
• viral infections yellow fever
influenza Measles SARS CoV2

249
Q

How are CD8 + responses induced?

A

induced by live attenuated microbes that can invade host cells

Usually very effective – excellent mimetics of the natural microbe

Risk is that they will still cause disease in some individuals e.g. immunocompromised
Activates

Another approach:
Injected DNA or mRNA is taken up by host cells
The protective sub-unit is produced by host cells and seen as foreign by the immune system
Protein antigen is processed via the MHC I (and MHC II) pathways.
Combine the advantages of live and sub-unit vaccines

Work well in animals not really humans

250
Q

Vector vaccine?

A

Inset gene encoding the protective subunit into a “harmless” microbe
• A wide range of carrier microbes have been suggested e.g.
– Disabled Salmonella – Lactococcus
– Vaccinia virus

251
Q

Polyclonal vs monoclonal antibodies?

A

Because most antigens carry many different epitopes,
animals injected with a single antigen will produce a
complex mixture of antibodies, each made by a
different clone of B cells

This antibody response is said to be polyclonal and
will not discriminate between different antigens

To discriminate between different antigens,
antibodies need to be identified that bind to epitopes
specific to individual antigens

These are known as monoclonal antibodies

252
Q

Do cancer cells or malignant cells have unique epitopes?

A

yes

253
Q

What is hybridoma technology?

A

Allows the identification and culture of cells
secreting identical antibodies with pre-defined
specificity

A single clone of cells secreting a single
antibody is made by fusing a B cell (Ig+ splenocyte
with finite lifespan) with a myeloma cell (cancerous
Ig- B cell with infinite lifespan)

The resulting hydrid cells (HYBRIDOMAS) each
make a specific monoclonal antibody

254
Q

Time course of a typical immune response when injected with antigen?

A

Primary injection:
Production of IgM
Multivalent
Low affinity

Secondary injection:
Production of IgG
Bivalent
Low to medium affinity

Tertiary injection:
Production of IgG
Bivalent
Medium to high affinity

Multiple boosts:
IgG
Bivalent
High affinity

255
Q

Make sure to look at photo library

A

ok

256
Q

Mechanism of hybrid cell selection in HAT medium?

A

HGPRT = Hypoxanthine:guanine phosphoribosyl transferase = blocks RNA and DNA transcription only can go through salvage pathway

Spleen cell = HGPRT+ but is not able to grow in long-term

Myeloma cell =HGPRT-, therefore not able to use the salvage pathway for RNA synthesis because the normal pathways are blocked

Hybridoma cell = has HGPRT from a spleen cell and immortality from a myeloma cell, therefore is able to grow in the HAT medium (providing there is an exogenous supply of Hypoxanthine and Thymidine)

257
Q

What is the PTA-ELISA?

A

Plate-Trapped-Antigen-Enzyme-Linked
Immunosorbent Assay

Find out antibody is present when substrate added for target antigen goes from purple to yellow

258
Q

What is phage display?

A

An issue with hybridoma technology is immuno-
dominance

Alternative technologies to generate mAbs include
phage display

A technique in which antibody fragments
are expressed as a fusion with a coat protein of a
bacteriophage

Fused proteins are displayed on the surface of the
virion, while the DNA encoding the fusion resides
within the virion

259
Q

Making antibody combinatorial libraries?

A

B cell source (from immunised animal)

extract the mRNA

reverse transcribe to get cDNA

PCR to amplify each of the V-gene families

Assesmble the VH and VL genes ar random (creates novel antibody repertoire with new pairings and specificities not present in donor)

Clone as scFv or F(ab) onto pIII or pVIII

260
Q

Describe the expression of scFv on the surface of phage?

A

scFv = VL and VH (V heavy and V light) domains of an immunoglobulin linked by a peptide spacer

Fusion proteins between scFv and pIII minor coat protein allows expression of scFv on surface of phage

Five copies of PIII expressed

Up to five scFv can be expressed on phage

261
Q

Describe the expression of F(ab) fragments on filamentous phage?

A

F(ab) fragments of antibodies can
be expressed on the surface of
phage as fusion proteins with pIII

Multiple copies expressed along the
length of the phage particle

262
Q

Uses of monoclonal antibodies?

A

Diagnostic pathology (cytology and histology)

In vitro diagnostic assays based on e.g. Enzyme-Linked Immunosorbent Assay (ELISA), Lateral-Flow Assay
(LFA) for pathogens and biomarkers of disease
(Lectures 15 & 16)

Affinity purification and characterisation of antigens

Limited use of mouse mAbs in vivo until relatively recently

263
Q

The need to humanise monoclonal antibodies?

A

Immunogenicity - most mAbs used in vivo are of mouse
origin and are therefore XENOGENEIC resulting in
Human Anti-Mouse Antibody (HAMA) reactions

  1. Origin - Mouse origin diminishes their ability to elicit effector mechanisms such as COMPLEMENT-MEDIATED LYSIS or ANTIBODY-DEPENDENT CELL-MEDIATED CYTOTOXICITY (ADCC)
264
Q

How does humanisation occur?

A

Use of xenoantibodies in human patients had
proved to be severely limiting owing to their
being seen as foreign proteins by the patient’s immune system

Strategies adopted to increase the human content of
antibodies which have proven clinical applications include:

Production of human hybridomas from human B lymphocytes

Replacement of the constant regions of mouse mAbs with those of human antibodies yielding CHIMERIC antibodies

Replacement of complementarity-determining regions (CDRs) of a human antibody with those of a mouse antibody via CDR GRAFTING to make a HUMANISED antibody

Production of FULLY HUMAN mAbs in transgenic mice

265
Q

Human immune system (HIS) mice?

A

CRISPR-Cas9 technology used to knockout Prkdc and IL2rg loci

Prkdc and IL2rg are part of the SCID (Severe Combined Immuno Deficiency) family of genes affecting maturation and formation of T cells, B cells, NK cells and other leukocytes

Knockout renders mice immunodeficient

Transplantation and engraftment of human HSC leads to mice with HIS

266
Q

Evidence for immunity and cancer surveillance?

A

Immunocompromised individuals more susceptible to cancer than general population

Some cytokines e.g. IL-2 are immuno-protective against cancer (IL-2 treatment is FDA approved for some cancers)

The tumour micro-environment is rich in Tumour Infiltrating
T-lymphocytes (TILs)

267
Q

The 3 phases of cancer immune surveillance?

A

Elimination:
By B cells, antibody, T cells, Treg cells, NK caells
This is the innate and adaptive immune response active against tumour
Can result in tumour cells death

Equilibrium:
Tumour is contained by the immune system but not completely destroyed

Escape:
Tumour antigen editing via genomic instability causes immune evasion
Tumour microenvironment becomes immunosuppressive

268
Q

The impediments to anti tumour immunity?

A

Immunocompromised individuals more susceptible to cancer than general population

Some cytokines e.g. IL-2 are immuno-protective against cancer (IL-2 treatment is FDA approved for some cancers)

The tumour micro-environment is rich in immune suppressive signals such as:

+ High levels of suppressive cytokines

+ Infiltration of Tregs (inhibit dendritic cells and therefore T cell activation)

+ Low MHC expression on tumour cells

+ High expression of immune checkpoint proteins on T cells

+ High expression of immune checkpoint protein inhibitors on tumour cells

269
Q

What does immunotherapy do?

A

Specific:
Trains the immune system to recognise and target only cancer cells

Powerful:
Attacks the cancer systemically through out the body

Universal:
A treatment approach that can be applied to nearly all cancers

Memory:
Capacity for memory means durability of protection

270
Q

Types of immunotherapy?

A

Passive vaccination (Therapeutic Monoclonal Antibodies)

Active vaccination e.g. HPV vaccine (Gardasil, Cervarix) and cervical cancer

Immune system modulators e.g. IL-2 (activates CD8 T cells)

Immune Checkpoint Modulation

Immune Cell Therapy

271
Q

How do therapeutic monoclonal antibodies target cancer?

A

Carry cytotoxic agents to tumour

Induce Complement-Mediated Lysis or ADCC of tumour cells e.g. Rituximab

Block activity of tumour-specific proteins e.g. Herceptin

Steps:
Naked MAb
Immunoconjugates
Multistep targeting

272
Q

What is non Hodgkins lymphoma?

A

Non-Hodgkin’s Lymphoma (NHL) is a malignant growth of
B cells that results in tumors in lymph nodes and
throughout the body

No cure for most patients, and only 60% of patients treated with
radiation and chemotherapy are still alive after 5 years

273
Q

What is Rituxan?

A

Rituximab is the first monoclonal antibody treatment approved for
cancer

Rituxan is a chimeric human-mouse mAb targeted against CD20,
an antigen present on the surface of neoplastic B cells

It consists of a human IgG1k antibody, with mouse variable
regions isolated from a murine anti-CD20 mAb, IDEC-2B8

In vitro studies show the mAb is able to lyse CD20+ cells using
human complement or ADCC

274
Q

Features of breast cancer?

A

Breast cancer is a common cancer and 1 in 9 women will develop it
during their lifetime. It also occurs in men.

The reasons for breast cancer are not well understood. Only 5-10% of
cases are thought to be inherited

Two breast cancer genes have been identified: BRCA1 and BRCA2

The most frequent mutations in BRCA1 - common in a single group, the
Ashkenazi Jews

Women with mutations in the BRCA2 tumor suppressor gene have a
greatly elevated risk of developing breast and ovarian cancers

Breast cancer cells express an antigen (protein) on
their surface called HER2

Breast cancer cells divide and grow when the
protein Human Epidermal Growth Factor attaches
itself to HER2

275
Q

What is Herceptin?

A

Herceptin is a humanised monoclonal antibody

It blocks attachment of human epidermal growth
factor to HER2

Herceptin is mainly used to treat women with
advanced breast cancer and is given in
combination with chemotherapy drugs such
as Taxol

After surgery, Herceptin treatment in combination
with chemotherapy halved the rate of recurrence

276
Q

Ahzheimers disease treatment called sola?

A

Solanezumab (Sola) neuroprotector

mAb binds to amyloid-b peptides

Binds to central epitope KLVFFAD
with picomolar affinity

Epitope is nucleation site for Ab
polymerization

Oligomers of Ab toxic to neurons

277
Q

Tocilizumab for rheumatoid arthritis?

A

Tocilizumab

Humanised anti - human IL - 6R receptor antibody

Blocks binding of IL - 6

278
Q

Mechanism of Immune Checkpoint Protein Inhibition?

A

Co-stimulation via CD28 ligation transducer T cell activating signals

CTLA-4 ligation on activated T cells down regulates T cell responses

Blocking CTLA-4 ligation enhances T cell responses

Immune Checkpoint Proteins (e.g. CTLA-4) keep immune responses in check by preventing overly intense responses that might damage normal cells

Blocking the activity of immune checkpoint proteins releases the “brakes” on the immune system, increasing its ability to destroy cancer cells

The first such drug to receive FDA approval, ipilimumab (Yervoy®), for the treatment of advanced melanoma, blocks the activity of a checkpoint protein CTLA-4 which is expressed on the surface of activated Cytotoxic T-Cell. This increases T-Cell activation.

279
Q

What is Nivolumab approved for?

A

approved for patients with melanoma, lung cancer, renal cell carcinoma, Hodgkin lymphoma, head and neck cancer, colon cancer, liver cancer

280
Q

What is pembrolizumab approved for?

A

approved for patients with melanoma, lung cancer, head and neck cancer, Hodgkin lymphoma and stomach cancer

281
Q

What is theralizumab - TGN1412?

A

Investigated immune modulatory drug for rheumatoid arthriris

Binds to CD28 co-stimulator pair on T cells

Potent agonist - stimulates anti inflammatory cytokine production

However caused severe inflammatory reactions due to cytokine release syndrome

282
Q

What is adoptive immune cell therapy: Normal TILs?

A

Idea behind this approach is that the TILs have already shown the ability to target tumor cells, but there may not be enough of them within the tumour to eradicate it or overcome the immune suppressive signals that are being released there

Introducing massive amounts of activated TILs can help to overcome these barriers and shrink or destroy tumors

In some experimental cases these treatment responses
have lasted for years

TILs are collected from samples of the tumor

TILs that show the greatest recognition of the patient’s tumor in lab are selected

Cells are then activated with cytokines and infused into the patient’s bloodstream

283
Q

What are transgenic TILs?

A

T cells are engineered to have a specific transgenic TCR which we know has high affinity to a certain tumour antigen

284
Q

What are CAR-T cells?

A

Another form of ACT that is being actively studied is Chimeric Antigen Receptor Modified T Cells (CAR-T cells)

A patient’s isolated T cells are collected from the blood and genetically modified to express a protein known as CAR

CARs are modified forms of a T cell receptor, which is expressed on the surface of T cells. These receptors allow the modified T cells to attach to specific proteins on the surface of cancer cells. Once bound to the cancer cells, the modified T cells become activated and attack the cancer cells

The modified cells are grown in the laboratory to produce large populations of the cells

CAR-T cells are then infused into the patient.

285
Q

Features T cell Receptors in treating a tumour cell?

A

Sensitive signal amplification derived by evolution

Low avidity

Targets intracellular proteome

Requires MHC class 1 expression and HLA matching on tumor cell

Possible misfiring with endogenous TCR

286
Q

Features of CAR targeting tumour cells?

A

Signal amplification derived by synthetic biology

Avidity controllable

Targets only surface structures

HLA independent antigen recognition

No misfiring with endogenous TCR

287
Q

What is antibody guided molecular imaging?

A
Radio-labeled antibodies used in nuclear medicine to locate
cancer cells (or infections) by molecular imaging using:

Computed Tomography (CT)
Magnetic Resonance Imaging (MRI)
Positron Emission Tomography (PET) - PET/CT or PET/MRI

Antibody Capromab reacts with prostate membrane specific
antigen (PMSA) on prostate cells

Can be conjugated to 111I with chelator pendetide and the radio-labeled antibody (trade name Prostascint) used to locate cancerous prostate cells. Others include antibody J591.

288
Q

Summary?

A

Monoclonal antibodies produced using hybridoma or phage display technologies are highly specific and can be used in immunotherapy and molecular imaging of cancers

Tumour associated antigens can be recognized by the immune system

Tumours adapt to evade immune clearance by mechanisms that either prevent recognition or that directly interfere with immune mediators

There at least 5 different modalities to circumvent this immune evasion

These modalities are collectively known as cancer immunotherapy

The most promising tools are immune checkpoint inhibitors and CAR-T T cells

289
Q

What is Ouchterlony immunodiffusion?

A

OID is a qualitative technique for detecting antibody-antigen
interactions in which antigens and antibodies are placed in a
gel of agar and allowed to diffuse towards one another

A positive reaction is indicated by the presence of PRECIPITIN lines due to a Antibody-antigen equivalence = immune complex formation

Antigen excess or antibody results in no precipitin

290
Q

Precipitin that forms when both antigen sources react with the antibody as they both have shared properties?

A

An arc

291
Q

Precipitin Line that forms when each antigen doesn’t share a common determinant of the antibody but both have a determinant of it?

A

A cross sword

292
Q

Precipitin line that forms when antigens share one common determinants but one of them has both required for the antibody

A

Arc on the side which doesn’t have an extra shared one

Spur forms on side that does, so cross bridge shape there

293
Q

What is Western blotting?

A

A technique in which specific antibodies are used to identify
particular antigens in mixtures of proteins that have been
resolved and transferred to a membrane

The proteins are separated by an appropriate technique,
usually SDS-PAGE (POLYACRYLAMIDE GEL ELECTROPHORESIS)

Transfer is carried out by electroblotting to a sheet of
nitrocellulose or PVDF, which is then incubated with labeled
antibodies specific for the antigen of interest

Radiolabeled or enzyme-linked antibodies can be used

Unbound antibodies are washed away, and the position of the
labeled antibodies is revealed using an appropriate technique

 Chemical substrates (Chemiluminescent or Colourimetric)
 Radiography
294
Q

Features of an SDS page?

A

The speed of migration in an electrical field depends on the dimension form and charge of molecules

For deaggregation and denaturation of the proteins SDS, B-mercaptoephanol or DTT (reducing agents) and heat is used

SDS provides negative charge to the proteins

Right to left = separated by charge by isoelectric focusing (IEF)

Up to down = separated by size by SDS electrophoresis

295
Q

What is immunofluorescence?

A

A qualitative technique in which antibodies tagged with
fluorescent reagents (Fluorescein IsoThioCyanate or
Texas Red) are used to detect molecules of interest.
FITC and TR fluoresce under UV at different absorbance
and emission spectra

IF is one of the most widely used techniques in immunology,
cell biology and clinical medicine

Used to localise antibodies or antigens on cell surfaces or
inside cells and to identify pathogenic organisms in tissues
or exudates

IF can be DIRECT or INDIRECT

296
Q

What is direct immunofluorescence?

A

Treatment on cell with labelled antibody

Unbound antibody washed away and UV light applied

Fluorescence observed where antigen is located

297
Q

What’s indirect immunofluorescence?

A

Treatment with unlabelled primary antibody

Unbound antibody washed away

Treatment with labelled secondary conjugate

Unbound conjugate antibody washed away UV light applied

Fluourescnce observed where antigen is located

298
Q

What’s immune-electron gold microscopy?

A

A qualitative technique in which antibodies tagged
with gold particles are used to detect molecules of
interest

EM-gold allows localisation of antigen-antibody
interactions at the nm scale (cf. IF at the m)

A powerful method of visualising intracellular antigens
and organelles

299
Q

What is Enzyme-linked immunosorbent assay?

A

ELISA is a quantitative technique for detecting antigens or
antibodies using enzyme-substrate reactions

The enzymes are usually coupled to antibodies either
DIRECTLY (to the antibody specific for the antigen) or to anti-immunoglobulins (INDIRECTLY)

The two main ELISA formats are:

  1. Plate-Trapped-Antigen (PTA)-ELISA
  2. Sandwich-ELISA

Quantification in ELISA is achieved using standard calibration curves

300
Q

4 types of plate ELISA?

A

Direct
Indirect
Sandwich ELISA
CompetitiveELISA

301
Q

Rhizoctonia solania case study?

A

Ubiquitous soil-borne plant pathogen with worldwide distribution and wide host range

Difficult to control using chemicals
Soil dwelling
Widespread resistance to fungicides
Soil fumigants are banned (Class II ozone depletors)

Most effective way to prevent disease is to ‘evade’ the pathogen altogether - test soils for infestation prior to sowing with susceptible crops

R. Solon-specific mAbs = Monoclonal antibodies raised against a surface glycoprotein antigen

mAb EH2 - protein epitope

mAb EE1 - carbohydrate epitope

Antigen is secreted by live cells only

Proces:

Stimulate active growth using baits

  1. Detect R. solani using specific mAbs
  2. Recover positive isolates for further testing
302
Q

Sandwich-ELISA format for R.solani?

A

Coat plates with mAb EH2 16h at 4 degrees

Bait incubation 2h and 23 degrees

add mAb EE1, 1h at 23 degrees

Anti-IgM (u-chain specific) peroxidase conjugate (1h at 23 degrees

Substrate formed 30min at 23 degrees

303
Q

What’s Batrachochytrium dendrobatidis

A

amphibian killer

Spores swim towards spogs

Burrow into them

Mature into sporangium

Spores released

Kills the frog within 120 days

Kills by colonising keratinised skin layer

Hyperplasia + Hyperkeratosis = disrupted electrolyte balance

Leads to cardiac arrest and death

304
Q

Features of Aspergillus disease?

A

Caused by Aspergillus fumigatus

Most important opportunistic mould pathogen of immuno-compromised humans esp. haematological malignancy and bone marrow transplant patients

Can result in -
Invasive Pulmonary Aspergillosis >300,000 cases/year worldwide with 30-95% mortality

Chronic Pulmonary Aspergillosis ~3 million cases worldwide in patients with underlying lung diseases including asthma

Allergic Bronchopulmonary Aspergillosis ~4 million cases worldwide in patients with asthma and cystic fibrosis

Covid-19-Associated Pulmonary Aspergillosis (CAPA) / IAPA?

305
Q

Diagnosis for Aspergillus infection?

A

Diagnosis notoriously difficult - patient history, culture from invasive biopsy, and biomarkers in invasive BALf or in blood

Hospital treatments ‘fever-driven’ or by non-specific chest CT - patients can receive inappropriate treatment with costly and toxic antifungal drugs

Imperative that diagnosis is made without delay - prognosis worsens rapidly over time with high mortality - exacerbated by poor diagnosis

Drug resistance in clinical strains of Aspergillus is increasing

306
Q

Mouse mAb JF5 for detecting Aspergillus?

A

IgG3 immunoglobulin

Recognises extracellular glycoprotein antigen

Antigen is secreted during active growth of hyphae
and is not produced by dead or dormant spores

JF5 used to develop a rapid, user-friendly, diagnostic test (lateral-flow assay) for detection of IPA using BALf and serum

307
Q

What is Bronchoalveolar lavage?

A

Lung fluid

308
Q

Commercialisation process of a monoclonal antibody lateral flow company?

A

Institute to industry

Patent to product

Bench to Bedside

309
Q

What’s non invasive antibody guided molecular imaging with immunoPET/MRI?

A

Can use it show up Aspergillus infection in a scan

It acts as a tracer

Depletion of neutrophil granulocytes in C57BL/6 mice via injection (i.p.) of the anti Gr-1 antibody RB6-8C5

Intratracheal (i.t.) infection of neutropenic C57BL/6 mice with A. fumigatus conidia

In vivo & ex vivo bio-distributions of [64Cu]-labeled antibody tracers measured in lungs via how much radioactivity there is

Next step of developing the technique -

Humanisation of JF5 by CDR grafting

Proved that the target epitope (b1,5-galactofuranose
) of the antigen is not present in humans