• 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

1/37

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;

37 Cards in this Set

  • Front
  • Back
What is immunologic tolerance?
Lack of response to antigens induced by exposure to these antigens
Central vs Peripheral Tolerance:
Location
Cells Involved
Deletion/Anergy
Central Tolerance: when developing lymphocytes encounter antigen in the primary (central) lymphoid organs: thymus, bone marrow

Lymphoid precursor exposed to self-antigen, if immature lymphocyte has receptor for self-Ag, it is deleted (undergoes apoptosis).

If not, maturation of clones not specific for self-Ag pass to peripheral lymphoid tissues.

Peripheral: when mature lymphocytes encounter antigen in the peripheral (secondary) lymphoid organs

Mature lymphocytes exposed to self-Ag, if responsive, cell undergoes apoptosis or anergy (non-reactive). If non-reactive, they wait for the right foreign Ag.
How is negative selection carried out? (Cells involved)
Negative Selection: When cells that react too strongly with Ag's are induced to undergo apoptosis

Double positive thymocyte is exposed to an APC with self-antigen; if the CD4+8+ cell exhibits strong recognition, it undergoes apoptosis
Function of AIRE?
AIRE = Autoimmune Regulator

Protein responsible for expression of peripheral proteins in thymus; increases exposure of developing T cells to normal proteins from throughout body

Facilitates T cell education

Note: All self-Ag's from throughout the body are NOT presented in thymus or BM (we don't just rely on Central Tolerance!)
What is anergy? When does it occur?
Functional inactivation of T cells

Occurs when Ag is recognized without co-stimulation
Where do Regulatory T Cells arise?
What differentiates them from other T cells?
What is their role? How is this carried out? (2 ways)
Tregs arise in thymus and peripheral lymphoid organs

Characterized by CD25 and FoxP3 expression

Ensure effector T cells aren't reacting to self-Ag

1) Contact Dependent Suppression:
CD25 will bind IL-12 being released by APC's (Naive T Cell won't bind it)

2) Cytokine-Mediated Suppression:
Treg (from thymus!) releases IL-10, TGF-beta which inhibits Th1 cells (prevents them from activating macs)
APC-Naive T Cell Signals Required for:
Clonal Expansion
Anergy (2 ways)
Clonal Expansion (Normal Response):
APC binds with MHC-Ag, B7 binds T cell's CD28

Anergy:
1)APC presents MHC-Ag, but not co-stimulation
2) APC presents MHC-Ag, B7 binds CLTA-4 receptor on T Cell
APC-Naive T Cell Signals Required for:
Clonal Expansion
Deletion (2 ways)
Clonal Expansion:
APC with MHC-Ag and co-stim binds Naive T Cell and releases IL-2-->Anti-apoptotic Protein produced-->CLONAL EXPANSION

Death Receptors:
APC with MHC-Ag, NO CO-STIM binds Act'd T Cell, IL-2 release-->Fas and FasL expression-->APOPTOSIS

Pro-Apoptotic Proteins:
APC with MHC-Ag, NO CO-STIM binds Naive T Cell, pro-apoptotic protein expressed-->APOPTOSIS
How do expression of CD8 T Cell populations and Fas-receptors correlate during the onset of adaptive immune response?
Slow, steady rise of CD-8's
Much more rapid Fas-receptor expression, begins decline when CD8 cells reach peak
Describe the molecular events following binding of FasL to FasR.
FasL binds FasR and forms FADD (Death Complex)
Stimulates Caspase 8
Cleaves Caspase 3
Cleaves PARP (DNA repair TF)
Cell Death
Describe the molecular events following IFN-alpha binding IFNR.
IFNalpha binds IFNR
Pi JAK
Pi STAT
Results in Gene Transcription
What is the effect of IFNalpha release on cell apoptosis?
IFNalpha inhibits Fas Signaling; it provides protection from apoptosis
Tolerogenic Self Antigens vs Immunogenic Foreign Antigens:
Presence in Generative Organs
Presentation with Second Signals
Persistence of Antigen
Self-Ag's:
Present in Primary Lymphoid to induce negative selection and Tregs

No co-stimulation; deficiency of second signals lead to anergy or apoptosis

Long-lived; repeated T cell activation induces apoptosis

Microbe:
Not presence in PRIMARY lymphoid organs; concentrated in PERIPHERAL lymphoid organs

Yes, microbes usually present with secondary sygnals to promote lymphocyte survival and activation

Usually short-lived bc immune system eliminates Ag
Where does B cell CENTRAL tolerance occur? How does it differ from T cell central tolerance?

Under what conditions would apoptosis be induced?
B cell Central Tolerance occurs in Bone Marrow

Differs because receptors can undergo editing when self-Ag is encountered

If immature B cell exhibited strong recognition of self-Ag, it'd undergo apoptosis
Where does B cell PERIPHERAL tolerance occur?

How are self-antigen recognizing cells weeded out? (2 ways)
Occurs in peripheral lymphoid tissues

Ag recognition without help!

If Mature B Lymphocyte recognizes a self-Ag it is inactivated (Anergy)

B cells that are partially activated by recognition of self-Ag's may be excluded from lymphoid follicles and die by apoptosis bc deprived of survival stimuli
How could self-reactive lymphocytes destroy tissue during time of infection?
Cytokines released cause influx of lymphocytes, activation of self-reactive lymphocytes, and subsequent tissue destruction (autoimmune disease)
Which gene mutations are most associated with self-reactive lymphocytes?

Give an example of such a gene and diseases its associated with.
MHC Genes: HLA alleles

HLA D4: Rheumatoid arthritis, IDDM, Pemphigus Vulgaris
Disease Association for C2, C4 (complement) mutations?
Lupus-like disease
Disease Association for Fas, FasL mutations?
Autoimmune Lymphoproliferative Syndrome (ALPS) via defective elimination of self-reactive T and B lymphocytes
Disease association for FoxP3 mutations?
Immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (also lymphoproliferative diseases) due to deficiency of Tregs
Disease Association for AIRE mutations?
Autoimmune Polyendocrinopathy due to defective elimination of self-reactive T cells in thymus (defect in central tolerance)
How is autoimmunity achieved by molecular mimicry?
Microbial antigen looks enough like self-antigen that when presented to Self-Reactive T cell (along with second signal), T cell becomes activated and destroys self tissue
How is autoimmunity achieved by induction of costimulators on APCs?
Microbes may activate the APCs to express costimulators(B7) and when these APCs present self-ag's, the self-reactive T cells are activated (B7 attaches to CD28 receptor), rather than being rendered tolerant.
In order to trigger immunity, tumor antigens must be recognized as non-self. List three ways this can be achieved.
Mutations
Over-expression
Aberrant expression (expressed at a location or time when they shouldn't be)
What is the effect of immunodeficiencies on tumor development?
Immunodeficient individuals have an increased incidence of some tumor types--suggests that immune system protects against growth of tumors (immune surveillance)
What is the effect of inflammation on tumor development during chronic infection? Provide 2 examples.
Chronic inflammation leads to tolerization of Ag's, which contributes to the development of cancer.

Ex: Hep B: hepatocellular carcinoma
H. pylori: gastric carcinoma
Ras, Bcr/Abl are examples of ______________
oncogene products recognized by CD8+ CTL's
2 examples of an oncogenic virus.
HPV
EBV
Are tumor antigens intracellular or extracellular?

Which class of MHC molecule are they presented on?
Intracellular

MHC I
How is induction of a CD8+ T Cell against tumors achieved?
Cross-priming (cross-presentation):
Tumor cells or antigens taken up by DC's, processed, presented to T cells
B7 costimulation provide second signal to CD8's
May also stimulate CD4+ T Cells which release cytokines for dx/dy of CD8's

Dx/Dy'd CTLs kill tumor cells without requirement for costimulation or T cell help (Effector Phase)
How does vaccination against tumor antigens enhance anti-tumor immunity?
Engages APC's
How does combining chemotherapy with immunotherapy enhance anti-tumor immunity?
Chemo will damage vasculature and improve access for immune cells
Inflammation will help engage APC's
Initial tumor cell death will 'feed' APC's
What are three ways tumor cells evade the immune system?
1) Antigen-loss variant of tumor cell (fails to produce tumor Ag)
2) Class I MHC-deficient tumor cell (mutations in MHC genes or genes needed for Ag processing)
3) Production of immunosuppressive proteins or expression of inhibitory cell surface proteins (inhibition of T cell activtn, ex: IL-10, TGF-beta)
What are three vaccination methods that result in enhanced anti-tumor immune response?
1) Vaccinate with tumor antigen-pulsed (incubated) DC

2) Vaccinate with DNA plasmid containing cDNA for tumor antigen to express antigen or transfect DC's

3) Vaccinate with tumor cell expressing costimulators (B7) or IL-12 (B7 will stimulate tumor-specific T cell, IL-2 will enhance proliferation and dx/dy of tumor-specific T cells)
Role of EGFR in Non-Small Cell Lung Cancer?
Epidermal GF Receptor expressed on NSCLC to help them multiply in body
EGFRBi vs EGFRBi-Armed ATC
EGFRBi: bispecific Ab made of two Ab's, anti-CD3 and anti-EGFR that are chemically joined together so can bind both T cells and EGFR+ cancer cells

EGFRBi-Armed ATC: activated T Cell coated with EGFRBi so they specifically target and kill EGFR+ tumor cells

(Note: ATC achieved by exposing T cells to IL-2)
What is leukapheresis?
Blood transfusion that only selects WBC's, returns all other cells to donor