• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/108

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

108 Cards in this Set

  • Front
  • Back
Definition of
TOLERANCE
lack of response to certain antigens

induced by exposure of lymphocytes to those antigens
CENTRAL TOLERANCE
developing T/B cells see self-antigens in primary lymphoid organs and develop tolerance to them
PERIPHERAL TOLERANCE
mature B/T cells see self antigens in secondary lymphoid tissues and develop tolerance to them
What proportion of the population has an autoimmune disease?
1-2%
2 main factors in development of autoimmune disease
inheritance of susceptibility genes
--> failure of self-tolerance

environmental triggers
--> self-reacting B/T cells
What type of lymphocyte controls almost all immune response to protein antigens?
CD4+ T cells
principal mechanism of central tolerance
negative selection
Autoimmune regulator (acronym)
AIRE
What is responsible for thymic expression of many self-protein antigens?
AIRE
APECED stands for...
Autoimmune polyendocrinopathy with candidiasis & ectodermal dysplasia
Etiology of APECED
mutations in aire gene
Development of Tregs
some immature T cells that recognize self-antigens in thymus develop into Tregs and enter peripheral tissues
Defective central tolerance development can lead to...
autoimmunity
2 mechanisms of peripheral T cell tolerance
mature T cells recognize self-antigens in peripheral tissues, --> anergy (functional inactivation) or death

self-reactive T cells suppressed by Tregs
ANERGY
functional inactivation of T cells
When does anergy occur?
when T cells recognize antigens without adequate levels of costimulators
What molecule do T cells express when they recognize self antigens?
CTLA-4
What kind of molecule is CTLA-4?
high affinity receptor for B7
What happens when T cell expressing CTLA-4 encounters self-antigen on APC?
CTLA-4 engages B7, which inactivates T cell
What is the ACTIVATING coreceptor on T cells?
CD28
What is the INACTIVATING coreceptor on T cells?
CTLA-4
ACTIVATION-INDUCED CELL DEATH
self-reactive T/B cells are deleted after repeated activation by self-antigen

or recognition of antigens without second signals
2 mechanisms of activation-induced cell death
(1) CD4+ T cells: repeated activataion leads to coexpression of Fas and FasL

(2) antigen recognition --> proapoptotic proteins in T cells, but only microbial antigens --> ANTIapoptotic proteins. Therefore, self-reactive T cells die.
Which cytokine potentiates Fas-mediated apoptosis?
IL-2
Function of Tregs
immunosuppression
Where do Tregs develop?
primary or secondary lymphoid tissues
usual combination of coreceptors on Tregs
CD25+ CD4+ T cells
?What is CD25
alpha chain on IL-2R
cytokines released by Tregs
TNF-beta

IL-10
Function of TGF-beta and IL-10
immunosuppressive:

block ativation of T/B cells and Mphages
Generation and functino of Tregs depend on what genetic factor
TF: FoxP3
Mutation in FoxP3 --> ?
autoimmune disease
2 options for immature B cells that interact strongly with self antigens in bone marrow
negative selection (--> death)

change receptor specificity (express new Ig light chain)
What happens to mature B cells that encounter high concentrations of self-antigens in peripheral tissue?
--> anergic and not respond to that self-antigen

follicle exclusion
Diseases associated with auto-antibodies (SLE, etc.) are caused by...
a defect in tolerance mechanisms of B cells and T-helper cells
Most important of multiple genes predisposing individual to autoimmune disease
MHC genes
HLA allele can _____ of disease, but does not _____ the disease
HLA allele can increase the RR of the disease, but it does NOT CAUSE the disease
Do most patients that inherit a disease-related allele develop the disease?
NO
2 reasons an HLA allele may be "bad"
ineffective at displaying self antigen (--> defect in negative selection)

peptide antigens presented fail to stimulate Tregs
General role of infection in development of autoimmune disease
infections may activate self-reactive T/B cells
infectious prodromes often precede what?
development of autoimmune disease
Mechanism by which infection stimulates development of autoimmune disease
infection in a tissue --> local, innate immune response --> increased expression of costimulators and cytokines by local APCs

activated tissue APCs stimulate self-reactive T cells that econcounter self-antigens
Short version of infection --> autoimmunity mechanism
infection can "break" T cell anergy and promote survival and proliferation of self-reactive T cells
MOLECULAR MIMICRY
some microbes produce peptide antigens that cross-react with self-antigens

s.t. immune responses against microbial peptides also attack self
Alternative mechanism by which infection triggers autoimmunity
infections may trigger release of antigens no usually seen by immune system

release may stimulate autoimmune reaction against the tissue from which they were released
IMMUNE SURVEILANCE
adaptive immune system prevents the outgrowth of transformed cells

or destory them before they become tumors
How may malignant cells signal immune system
molecules recognized as foreign antigens

normal proteins overexpressed or dysregulated
Example of a self protein that, expressed on a malignant cell, may trigger immune response
protein found in embryonic but not adult tissue may not induce tolerance
principal immune mechanism against tumors
CTLS specific for tumor antigens
most tumor antigens are...
endogenously synthesized, cytosolic proteins
How are tumor antigens displayed to CTLs
by MHC I on APCs
From which types of cells can tumors arise?
any nucleated cell type
CROSS-PRESENTATION allows...
cells that don't express costimulators or MHC II to activate CD8+ T cells
CROSS-PRESENTATION mechanism
tumor cells ingested by APCs

APCs provide 2nd signals (costimulators) to activate T cells
Definition of CROSS PRESENTATION
One cell type presents antigens of another cell type

and primes T cells for 2nd cell type
After differentiation, CTLs can kill tumor cells with _____, but without...
with relevant antigens

without costimulators or T-helper cells
2 passive evasion mechanisms of tumors
growth can outstrip immune defenses

many tumor antigens are only weakly immunogenic because they differ so slightly from self-antigens
2 active evasion mechanisms of tumors
tumors stop expressing antigens that are the targests of immune attack

tumors stop expressing MHC I
Antigen-loss variants
tumors that stop expressing the antigens that are the targets of immune attack
Consequences of tumor stopping expression of MHC I
stop CD8+ T cell activation, but may be attacked by NK cells
3 main immuno-based anti-tumor strategies
provide anti-tumor effectors (Ig's and T cells)

actively immunize patients against tumors

stimulate patients own anti-tumor immune responses
HER2/neu
antigen overexpressed in patients with certain types of BRCA
immunological treatment for B-cell malignancies
mAb against CD20, expressed by B cells

CD20 not expressed by hematopoietic stem cells, so normal B cells replenished after treatment stopped
adoptic cellular immunotherapy
T cells isolated from blood/tumor infiltrates of patient

expanded w/ GFs in vitro

injected back into patient
3 means of increasing host's own anti-tumor responses
(1) vaccinate patients with own tumor cells/antigens - recominant proteins with adjuvants

(2) grow DCs & expose them them to tumor cells/antigens = "tumor pulsing" --> vaccine mimicking normal cross-presentation pathway

(3) vaccinate with plasmid of cDNA encoding tumor Ag - host cells, including APCs take up plasmid, express Ag, elicit specfic T cell response
Aim of anti-tumor treatment with cytokines, e.g.: IL-2
let patients generate own anti-tumor responses
limitations of IL-2 treatment for tumors
serious toxic effects
syngeneic
animals that are genetically identical (MHC alleles)
allogenic
animals that are not genetically identical (MHC alleles)
xenogenic
animals from different species
allografts and xenografts are always....
rejected
clinically, most transplants are...
(syngeneic/allogenic/xenogenic)
allogenic
all ____ molecules are potential targets for rejection
MHC
why do MHC molecules from another individual's cells elicit such a strong immune response?
cross-reaction:
allogenic MHC with allogenic-peptides look like self-MHC molecules with bound foreign peptides
minor histocompatibility antigens
non-MHC antigens that elicit graft rejection
minor histocompatability antigens usually take the form of...
allelic forms of normal cell proteins that differ between donor and host
which 2 transplant types are most affected by MINOR histocompatibility antigens?
blood transfusion

BM transplant
direct allorecognition
host T cell recognize unprocessed allogenic MHC molecules on graft APCs
indirect allorecognition
host T cell recognizes a processed peptide of allogenic MHC bound to self MHC on host APC
MLC
mixed lymphocyte reaction
definition of MLC
in vitro model of T cell recognition of alloantigens

T cells from 2 individuals cultured together

level of T cell response assayed is propotional to the extent of MHC differences
3 classifications of graft rejection
hyperacute

acute

chronic
timeframe for hyperacute rejection
within minutes
physiologic effects of hyperacute rejection
thrombosis of graft vessels

ischaemic necrosis of graft tissue
hyperacute rejection is mediated by...
circulating antibodies, specific for antigens on graft endothelium

these antibodies are present pre-transplant, possibly b/c of previous transfusions and reactions against alloantigens in transfused blood cells
pathways activated by circulating antibodies in hyperacute rejection
complement

inflammation

thrombosis
why is hyperacute rejection fairly uncommon?
clinical practice of cross-matching
time frame for acute rejection
days/weeks
principal cause of early graft failure
acute rejection
acute rejection is primarily mediated by which cell type?
CD8+ T cells
Effects of Ig's in acute rejection
contribute primary to vascular damage:

parenchymal cell damage

insterstitial inflammation

endothelialitis
time frame for chronic rejection
months to years
definition of chronic rejection
progressive loss of graft function
physiologic effects of chronic rejection
fibrosis of graft

gradual narrowing of vessels (graft atherosclerosis)
Effect of T cells in chronic rejection
secrete cytokines

stimulate fibroblasts and vascular smooth muscle cells in graft
mainstay of prevention/treatment of graft rejection
immunosuppression

designed to inhibit T cell activation & effector function
most useful drug for prevention of graft rejection
cyclosporine
mechanism of cyclosporine
blocks T cell phosphatase required to activate NFAT

thereby inhibiting cytokine genes in T cells
types of transplants allowed by cyclosporine
heart, liver, lung
problem with all transplant drugs
non-specific immunosuppression
transplant drugs increase the incidence of which 2 things?
infections

cancer
Is HLA matching always necessary?
No - b/c of cyclosporine
xenotransplantation elicits...
hyperacute rejection
describe antibodies that mediate hyperacute rejection in cases of xenotransplanation
not require previous exposure

may be produced against bacteria that normally inhabit gut & cross react with cells of other species
blood group antigens are which type of macromolecule?

which means they elicit which type of immune response
sugar

T cell response
Why does BM transplant in particular weaken the immune system
before transplant, part of host BM is destroyed to make room for graft
GVHD
graft-versus-host disease
etiology of GVHD
If mature T cells are transplanted with BM cells, can attack host tissues