• 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

1/167

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;

167 Cards in this Set

  • Front
  • Back
What are PAMPs?
Pathogen-associated Molecular Patterns
Examples of PAMPs?
LPS (lipopoly saccharide); Flagellin
Where do dendritic cells come from?
Mature from monocyte precursors
In what region of lymph nodes are B cells found?
Follicles
In what region of lymph nodes are T cells found?
Paracortex
What are M cells?
Multifenestrated cells; present in lining of small intesting -- form membrane overlying lymphoid compartments within a Peyer's patch; transport antigens from lumen of gut to APCs
What are the components of the innate immunity system?
Physical barriers, complement system, phagocytes, natural killer cells
What are the three major physical barriers in the innate immune system?
Skin, GI tract, respiratory tract
Where do macrophages come from?
Monocyte precursors that migrate into tissues
What is the process by which leukocytes are recruited to the site of an infection?
Macrophages secrete TNF and IL1 etc., causing endothelial cells to produce other proteins that make rolling leukocytes stick
What is the role of cytokines in innate immunity?
Mediate the early inflammatory reactions to microbes and promote elimination of microbes
Are cytokines used in innate or adaptive response?
BOTH
What is the role of cytokines in adaptive response?
Stimulate proliferation and differentiation of antigen-stimulated lymphocytes and activate specialized effector cells
What is chronic granulomatous disease?
Phagocytes lack the oxidases necessary to kill ingested gram + bacteria
What cytokines are involved in innate immunity?
IL-12 and IFN-γ
What is ADCC?
Antibody dependent cytotoxicity; how natural killer cells and eosinophils are signaled to lyse cells; involves recognition of Fc regions of bound antibodies by NK or eosinophils
What class of immunoglobin signals NK cells?
IgG
What class of immunoglobin signals eosinophils?
IgE
What do NK cells secrete as a signaling molecule? What cells respond to it?
IFN-γ; activates macrophages
What do NK cells respond to?
IL-12
What is the purpose of NK cells?
Eliminate reservoirs of infection
How do NK cells kill their targets?
Release perforin
How are normal cells spared from NK action?
MHC class I inhibits NK cell action
What are TLRs?
Toll-like receptors;serve as pattern recognition receptors for cells of innate immune system
What recognizes PAMPs?
TLRs
How does an innate immune response lead to an adaptive immune response?
Macrophages and dendritic cells express costimulators and secrete cytokinds that together activate T lymphocytes (signals 2 and 3)
What is the basic function of a helper T lymphocyte?
Activate macrophages and B cells
What is the basic function of a Cytolytic T lymphocyte?
Kill infected cells and eliminate reservoirs of infection
What is the basic function of a B lymphocyte?
Secrete antibodies that block infections and promote elimination of extracellular microbes
What are the three pathways of complement activation?
Classical, Alternative, MBL
What is the hallmark of the classical complement pathway?
Triggered by antibodies that bind pathogens (ie part of adaptive response)
What is the part of the complement system that promotes obsonization?
C3b binding (also coated with C4d, iC3b, and C3dg)
What is the function of the MAC complex formed by the late components of complement?
Osmotic lysis of the microbe
What initiates the classical complement cascade?
C1 must bind two or more Fc portions of IgM or IgG that are bound to antigen
What are the three responses that complement triggers?
Lysis, Phagocytosis, and Inflammation
What is the first common step of all three complement pathways?
Production of C3b
What component of complement stimulates inflammation?
C5a (also C4a and C3a)
How are immune complexes (opsonized antigen-antibody complexes) cleared from bloodstream?
Erythrocytes bind the complexes and carry then to spleen or liver where the complexes are cleared by the resident phagocytic cells (eg Kupffer cells in liver)
What is the effect of attaching C3d to microbial antigens in a nonimmune host?
Amplifies T-dependent B cell response to that antigen
What is the order of activation of the components of the classical complement pathway?
C1, C4, C2, C3, C5, C6, C7, C8, C9
How is the alternative complement pathway activated?
C3b binding to various activating surfaces (note that C3b is constantly being produced as part of innate immune response)
How is the lectin complement pathway activated?
Activates C4 in absence of antibody, then follows classical pathway; triggered by binding mannose on pathogen surfaces
What forms the MAC?
Multiple C9 molecules
What inhibits C1 activation?
C1INH-
What inhibits the convertases involved in complement?
DAF and Factor1
What inhibits the MAC
CD59 and S protein
What is angioneurotic edema?
Hereditary failure in regulation of C1 activity by C1 INH-
What is paroxysmal nocturnal hemoglobinuria?
Absense of functional DAF on erythrocytes (normally inhibits formation of C3 convertases)
What defects in complement lead to hemolytic anemia and strokes?
Deficiency of CD59, which normally inhibits formation of the MAC
What structural difference is there between IgM and IgG?
IgM has one more heavy chain C region domain; has C-terminal transmembrane and cytoplasmic portions that anchor molecule in plasma membrane
Where do the highly variable regions of antibody molecules map to?
The parts of the antigen binding surface loops (CDR1, CDR2, CDR3) that make the most contacts with antigen
What is CDR?
"Complementarity determining regions" -- highly variable regions of antibody molecules that make the most contacts with antigen
What proteins mediate recombination of antibody molecules?
RAG1 and RAG2 ("recombinase activating gene")
How many chromosomes are used to make antibody molecules?
Only One!
Genetic difference between heavy and light chain recombination?
Light chain gnes do not have D segments; two separate light chain loci (only one for heavy chain)
What is junctional diversity?
Result of inexact RAG-mediated cutting -- leads to different DNA sequences
About how many different antigen combining regions can be made?
10E23!
What is the process by which additional variability is introduced into IgV regions, and is associated with increased affinity for the antigen?
Somatic Hypermutation
Where is AID expressed?
Only in B cells
What is necessary for somatic hypermutation?
AID protein (specific for B cells)
What processes does AID mediate?
somatic hypermutation and immunoglobulin heavy chain isotype switching
What form of Ig is expressed ona Pre-B cell?
cytoplasmic mu and pre-B receptor-associated mu
What form of Ig is expressed on an immature B cell?
Membrane IgM (mu and kappa or lambda light chain)
What form of Ig is expressed on a Pro-B cell?
none
What form of Ig is expressed on a mature B cell (not activated)?
Membrane IgM or IgM and IgD
What provides second signal to B cells?
CD4+ T cells
Where does affinity maturation occur?
Germinal centers of follicles
What signals drive class switching of B cells?
CD40-CD40L and cytokines
[go through class switching and antigen recombination in book!]
Are RAG proteins involved in class switching?
No
What is the effector function of IgM?
Activation of classical complement pathway
What is the effector function of IgG?
Neutralization of microbes and toxins; opsonization to promote phagocytosis; activation of classical complement pathway; Feedback inhibition of B cell activation
What is the effector function of IgE?
Immunity against helminths (activation of eosinophils); Mast cell degranulation
What is the effector function of IgA?
Mucosal immunity (transport of IgA through epithelia)
What part of antibody structure changes during isotype switching?
C regions
What part of antibody structure changes during affinity maturation?
V regions
Where do naïve T cells preferentially migrate to?
Lymph nodes
Where do newly activated T cells preferentially migrate to?
Circulation
Where do effector and memory T cells in blood preferentially move to?
Peripheral tissues
[look up terminal deoxyribonucleotide transferase]
How does AID promote somatic hypermutation?
Converts C nucleotides to uridine (U), causing mutation by base pair substitution
What causes IgM and IgD to be co-expressed?
No stop site is present between mu and delta regions
What form of Ig is never secreted?
IgD
What triggers affinity maturation and class switching on B cells?
CD40-CD40L interaction between B and T cells
What cells can function as APCs?
Macrophages, dendritic cells, B cells
What are the MOST efficient APCs?
Dendritic cells
What is an example of an "MHC-like" molecule?
CD1; presents lipid antigens to unique population of lymphocytes
What cells recognice MHC class I molecules?
CD8+ T cells (CTLs)
What are the alleles for the MHC class I molecules?
HLA-A, B, C
Which MHC molecules are expressed on all nucleated cells?
Class I
what are the components of a MHC class I molecule?
single polymorphic heavy (alpha) chain and non-MHC encoded, non-polymorphic light chain called beta-2-microglobulin
What are the components of a MHC class II molecule?
Alpha and beta chains, both polymorphic
What cells express MHC class II?
APCs and thymic epithelial cells
What are the alleles for class II MHC?
DP, DQ, DR
What is another name for helper T cells?
CD4+ T cells
Peptides that end up in the cleft of MHC class I generally arise from:
proteins that are synthesized in the cell (can also contain peptides from proteins that escaped phagosomes)
Peptides that end up in the cleft of MHC class II generally arise from:
proteins that are made externally to the cell then taken up by professional APCs
What is the class II invariant chain protein?
Protein that stabilizes a newly synthesized class II MHC molecule before it fuses with an endosome containing broken down peptides from ingested antigens
What are TAPs?
The special transporters that transport peptide fragments into the ER where they bind to newly synthesized class I MHC molecules
How do DCs take up material that they will then present to T cells?
Macropinocytosis
What is DM protein? (involved with class II MHC proteins
present in endosomes that fuse with vesicles containing just-synthesized MHC class II molecules; competes for binding with invariant chain, freeing up binding site for antigenic peptide
What is the result of a TAP deficiency?
Variant of SCID; No peptides present in ER for class I MHC to bind
What is bare lymphocyte syndrome?
Variant of SCID; failed expression of MHC class II molecules
What is MHC restriction?
Idea that T cells are specific to self MHC molecules
How can you distinguish between T cells, B cells, and NK cells (in a lab)?
Cell surface markers stained by monoclonal antibodies
What are the two chains of the TCR?
alpha and beta
What gene segments are combined for the alpha chain of the TCR?
V, J, and C
What gene segments are combined for the beta chain of the TCR?
V, D, J, and C
What are the two chains of the BCR?
heavy and light
Major difference in binding between TCR and BCR?
TCR binds to peptide+MHC, and requires simultaneous binding of CD4 or CD8 to MHC; (BCR binds to just peptide, no requirement for simultaneous binding)
What is the alternate form of the TCR expressed by a minority of T cells?
gamma/delta; often responds to lipid instead of peptide antigens
What are Natural Killer T cells? What is their function?
Cells that have the surface markers of both T cells and NK cells; Critical for protection against autoimmunity and cancer
What are CD3 chains?
Series of proteins found in association with TCRs; responsible for initiating signal transduction
What is responsible for initiating signal transduction (activating the cell) after a T cell binds to MHC+peptide?
CD3 chains and CD4
What molecules play the role of co-stimulation (signal 2) for T cells being activated by APCs?
CD28 is receptor, B7 is ligand (on activated APCs)
What is necessary for T cell activation?
MHC-TCR interaction PLUS B7-CD28 interaction
What is the role of CD40/CD40L?
Activated T cells upregulate expression of CD40L; When this interacts with CD40 on an APC, that APC becomes activated to express more B7
What is CTLA4?
Molecule on T cells that is expresed late in the activation process; Binds B7 more avidly than does CD28, thus preventing CD28 signaling and terminating the T cell response (makes cell anergic)
What is the ITAM? (relates to T cell activation)
Specific domain within CD3 proteins that is responsible for initiating signal pathways
What is IL-2?
Interleukin-2; produced by activated T cells; promotes proliferation of T cells
How does TCR stimulation affect IL-2 interactions?
Stimulation causes production of IL-2 as well as expression of alpha chain of receptor, which makes receptor high-affinity; signaling results in cellular proliferation
What markers do immature T cells entering the thymus express?
None; negative for CD3, CD4, and CD8
How does homing of T cells occur?
Naïve T cells express L-selectin, which targets them to lymph nodes; after activation, E and P selectins are expressed, which target the cells to the site of infection
How do CTLs effect death of target cells?
Release granules containing perforin and granzymes (initate apoptosis) into target cells when restimulated by MHC class I presenting the antigen to which they are primed
What is anergy?
The state in which a lymphocyte has lost the ability to be stimulated; produced by receiving signal 1 without signal 2
What is the principal action of IL-4?
B cell switching to IgE
What is the principal action of IL-5?
Activation of eosinophils
What is the principal action of IFN-gamma?
Activation of macrophages
What is the principal action of TGF-beta?
Inhibition of T cell activation
If a naïve T cell is stimulated in the absence of TGF-beta but the presence of IFN-gamma and/or IL-12, what type of effector cells will develop?
Th1
If a naïve T cell is stimulated in the absence of TGF-beta but the presence of IL-4, what type of effector cells will develop?
Th2
If a naïve T cell is stimulated in the presence of TGF-beta and IL-6, what type of effector cells will develop?
Th17
If a naïve T cell is stimulated in the presence of TGF-beta and in the absence of IL-6, what type of effector cells will develop?
Treg cells
What is the dominant transcription factor for Th1 cells?
T-bet
What is the dominant transcription factor for Th2 cells?
Gata-3
What is the dominant transcription factor for Th17 cells?
ROR-gamma
What is the dominant transcription factor for Treg cells?
FoxP3
What do Th1 cells do?
activate macrophages to kill engulfed bacteria
What do Th2 cells do?
stimulate B cells to make antibody and class switch
What are the major cytokines produced by Th1 cells?
IFN-gamma and IL-2
What are the major cytokines produced by Th2 cells?
IL-4 and IL-5
Imbalance in what T cell effector group often leads to inflammatory diseases?
Th1
Imbalance in what T cell effector group often leads to allergic responses?
Th2
What is the major cytokine produced by Th17 cells?
IL-6 and IL-17
What seems to result from uncontrolled activity of Th17 cells?
autoimmunity
What do Th17 cells do?
Stimulate neutrophil recruitment
Note that CD4+ T cell subsets negatively regulate each other
What type of T cells can be subdivided into several subgroups based on the cytokines they produce?
CD4+ T cells
What is the difference between long-lived plasma cells and memory cells?
Memory cells can be any type of T or B cells; do NOT secrete anything. Plasma cells are B cells that continue to secrete small amounts of antibodies
How is apoptosis stimulated in T cells?
Engagement of Fas (CD95) receptor on activated T cells signals for apoptosis
What is AIRE?
Transcription factor involved in expression of self-antigens in thymus; genetic abnormalities result in auto-immune disease
What is APECED?
Autoimmune polyendocrinopathy with candidiasis and ectodermal dysplasia; caused by genetic abnormality in AIRE gene
In a primary immunodeficiency, upper and lower resp infections indicate what kind of deficiency?
B cell
In a primary immunodeficiency, obligate intracellular organisms indicate what kind of deficiency?
T cell
In a primary immunodeficiency, unusual bacteria indicate what kind of deficiency?
Neutrophils
If your IgG is less than 100mg/dl and you have no IgA or IgM, what do you have?
X-linked Agammaglobulinemia
If you don't have tonsils because of a congenital abnormality, what do you have?
X-linked Agammaglobulinemia
Chronic Granulomatous Disease is a disorder of what?
The INNATE immune system; can't make ROS to kill engulfed bad stuff in phagocytes
What is a type I hypersensitivity?
Allergies… IgE mediated; over stimulation of mast cells
What is a type II hypersensitivity?
Antibody mediated but not IgE; tissue damage and inflammation
What is a type III hypersensitivity?
immune complexes get deposited in blood vessels, cause inflammation
What is a type IV hypersensitivity?
T cell dependent
What type of antibody dominates in a secondary response (as compared with a primary response)?
IgG
What are the advantages and disadvantages of live attenuated virus vaccines?
Good immunogens that induce long-lied, appropriate immunity BUT are biochemically and genetically unstable and often not possible to produce; dangesrs of contamination or reversion to virulence
What are the advantages and disadvantages of inactivated vaccines?
Stable with little or no risk of virulence BUT not possible for all viruses and not as effective as live attenuated; does not create mucosal or cell immunity and often need reimmunization
What are the advantages and disadvantages of using fragments or sub-unit vaccines?
Safe BUT poor antigenicity and problems with vaccine delivery into body
What cells are responsible for tumor immunity?
T cells