Shared Flashcard Set

Details

Immunology Spring Exam 1
MWU immunology Spring Exam 1
97
Immunology
Graduate
03/23/2012

Additional Immunology Flashcards

 


 

Cards

Term
Hematopoiesis
Definition
The formation AND differentiation of BCs (WBC, RBC, Platelets) usually takes place in marrow of the ribs, sternum, and hips
Term
HSC
Definition
-precursor to all blood cells
-Pluripotent (can differentiate into multiple types of cells)
-Once they divide the daughters can either self-renew or proceed to on eof the lineages.
-<.1% of all cells in MARROW
Term
Cytokines
Definition
Chemical messengers produced by BONE MARROW STROMAL CELLS and dictate which lineage the HSC differentiates into
Term
Two main Blood Cell lineages, cytokines
Definition
Myeloid (IL-3, GM-CSF) Mono, Macro, Granulo, RBC, Platelets
Lymphoid (IL-3,7)- T,B,NK cells
Term
Constitutive differentiation
Definition
process that is happening all the time and continuously in the Red bone marrow to maintain basal levels
Term
Inducible hematopoieisis
What gives off cytokines?
Definition
reaction to something, hemmorrhage, injury, etc. Need more WBC so can be induced. Activated T-Helper and macrophages produce cytokines
Term
CD marker for HSC
Definition
34+
Term
Monocyte General characteristics
Definition
CD14
-Horseshoe shaped nucleus
-lots of lysosomes and phagocytic vacuoles
-LPS- component for gram negative bac.
-Cytokine producing, phagocytic, APC
Term
Macrophage General Characteristics
Definition
Come from monocytes- have moved into the tissues and differentiated
-RES formed by Macrophages
-Phagocytosis, APC, cytokine secretion for induceable hematopoiesis
Term
RES
Definition
Reticluo Endothelial System- network of macrophages throughout the body
Term
Dendritic Cell Classes and General Characteristics
Definition
APC- Proffessional APC
- Capture antigen and move to lymphoid organ to present to T cell
-From the myeloid lineage w/monocyte precursor
-long filamentous projections
-Langerhans and interstitial DC
2. Follicular Dendritic cells- aid in the differentiation of B cells, only in lymphatic organs
Term
Granulocyte General Characteristics
Definition
from the myeloid lineage,
Contain cytoplasmic granules
include Baso, Eosin, Neutrophils
Term
Neutrophil General Characteristics
Definition
70% of nucleated BCs
-released from bone marrow- migrate to tissue- first at site of inflammation
-first line of defense in microbial infection, bacteria, fungal
-phagocytosis (O2 dep/indep)
-very segemented nucleus
Term
Eiosinophil General Characteristics
Definition
2-5% of nucleated BCs
-biloped nucleus
-Stains red w/acidic dye
-defend against parasytic organisms via degranuliation, minor phagocytic role of particles that are broken up.
Term
Eiosinophil granules contain what?
Definition
MBP, ECP, EPO
Term
Basophil General Characteristics
Definition
Only found in Circulating Blood
-NONphagocytic granulocyte, stains purple
-Surface receptors for IgE and C3a
-Release granules containing histamine (important role in allergic response)
Term
Mast Cell General Characteristics
Definition
-Very similar to basophil except only found in the TISSUES
-From the myeloid lineage
-histamine granules and surface receptors for IgE and C3a
-role in allergic response
Term
Lymphocytes Basic info
Definition
20-30% of peripheral leukocytes
-in blood, tissues and lymphoid organs
-Small= T,B lymphocyte (high nuclear/cyto ratio)
-Large= Activated T cells, Plasma cells (activated B cells) or NK cells (only NK is non-activated large cell)
Term
Memory cell general Char.
Definition
long lived lymphocyte that patrol the body in defense of the pathogen ino order to mount a quicker defense in the future
Term
BLympho. gen. Char.
Definition
5-15% of lymphocytes
-differentiate and mature in Bone marrow.(Ca2+ important for maturation)
-BCR, CD19, CD20, CD21
-synthesize and secrete specifica antibodies to defened against extracellular microbes and toxins.
Term
TLympho gen char
Definition
80% of lympho.
TCR and CD3 on ALL T Cells
-all activated upon antigen exposure
-become effector or memory T cells
-Generated in bone marrow, migrate to Thymus (10% survive)
-Become Helper T cells, Regulatory cells, Cytotoxic T cells
Term
Helper T cells
Definition
-Helper T cells have TH or CD4+
-cytokine production to activate macrophages, induce inflammation, and induce proliferation and differentiation of T,B lymphocytes
Term
Regulatory T cells
Definition
Treg, CD4, CD25+
-cytokine production to controle autoimmune and turn off immune response
Term
Cytotoxic T Cells
Definition
Tc, CTLs, CD8+
-direct killing of tumor cells and cells infected with intracellular microbes
Term
NK Cells
Definition
Specific Antigens
-CD16a, CD57
-destroy tumor cells or virally-infected cells that have decreased expression of MHC class 1 proteins
Term
Neutropenia
Definition
low number, <1000 cells/ul
indicates increased risk of infection (esp from gram- bac)
Term
Neutrophilia
Definition
PMN (neutrophil)count >8000/ul
clinical sign of infection
-response of the bone marrow to circulating cytokines (left shift of band cells, immiture neutrophils)
Term
Eosinophelia
Definition
>600/ul
-due to parasitic infection, addison's disease, allergy, asthma
Term
BLympho and Epstein Barr Virus
Definition
Human Herpes Virus- exploits CD21
-Heterophile antibodies produced
Term
TH and HIV
Definition
TH cell is the target of HIV- High levels fo CD4 which HIV binds and uses to infect T helper cell
Term
Primary lymphoid organ
Definition
Bone marrow and thymus
-where lymphocytes mature
Term
Secondary Lymph Organ
Definition
Lymph nodes, spleen, MALT, Tonsils
-Where actual immune response takes place and lymphocytes respond to the antigen
-environment where antigen filter and lymphocytes circle throug to increase changes of interactions
Term
Bone Marrow Function
Definition
Hematopoiesis, Myeloid cells mature, b-cells formed and mature, T cells formed and move on
Term
Thymus function
Definition
location of t-cell maturation into three types that are aducated about self and non-self
-size and function decreases with age (dec in t-helper and Regulatory T cells)
Term
Lymph node
Definition
secondary lymph organ
-B cells in cortex and T-cells in paracortex b/c activated differently
-filter antigen from local tissue via afferent lymphatic vessels or DC cells- site of lymphocyte activation.
-B and T cells circulate through and only stay if they see the antigen (takes 7-10 days to be activated) or move through to next node if don't see one.
Term
Lymphatics
Definition
network of capillaries and vessels that transports lymphocyte and antigesnf rom body tissues to lymph nodes.
-fluid moves as a result of muscle contraction
Term
Spleen
Definition
secondary lymph organ
-filters angigen from the blood adn site of lymphocyte activation
Red Pulp- destroy old and damaged RBCs via phagocytosis
Wite Pulp- lymph tissue- antigens enter via the splenic A and exit via the splenic A
B cells in Follicles
T cells in PALS
Term
MALT
Definition
large pop of antibody producing plasma cells
-loose arrangement of cells and connective tissue that defends mucus-lined epithelial cells
-majority of antigens come from MALT due to the large size.
-BALT or GALT depending on location
Term
Tonsils
Definition
Oral, nasal and paryngeal tissue
-Partially encapsulated MALT
Term
Extravasation
Definition
movement through an unruptured vessell into surrounding tissues
-CAMS slow it down so it can get through the junctions
Term
CAMS
Definition
molecules on endothelium that slow leukocytes so they can enter tissues (extravasate)
Selectins- form a loose interaction that slows the WBC down (rolling)
Integrins- stronger bond that actually stops the cell so that it can move through.
Term
Chemotaxis
Definition
Directed movemnt through tissue eliicited by chemokines and chemokine receptors on cells
-ex. IL-8 attracts Neutrophils
Term
Review recirculation patterns for Naive lymphocytes
Definition
Review recirculation patterns of Activated lymphocytes (really do this!)
Term
Self
Definition
Normal body constituents or natural components of an individual
Term
Non-Self
Definition
Cells or Molecules to which immune system has no exposure during its maturation ro does not normally encounter in the body
-Threats= antigens, viruses, bacteria, parasitic organisms, toxins, transplanted tissues, cancer (if a cell responds to self during maturation then it is selected against)
Term
Characteristics of innnate immunity
Definition
born with all components
1. Structural- TJs Mucus Cilia Sweat Tears Saliva
2. Chemicals- Digestive enzymes acids lysosomes defensins (embed into membrane of bacteria, create pores and destroy)
3. Phagocytes- Neutrophils, macrophages, monocytes, DC
4. NKs
5. Granulocytes
6. Complement system
-These all have the same mechanism regardless of antigen and the response is in place before the threat is encountered.
-no memory is generated, doesn't enhance or change w/ inc. exposure
Term
Characteristics of adaptive immunity
Definition
T and B cells respond very specifically to a specific antigen.
1.Memory-develops after a first exposure of B and T cells to a specific antigen (activation is much quicker, repeated exposure increases response)
2.Specificity- Each T,B cell has surface receptors that are speficic for one antigen only.
3.Humoral
4.Cell Mediated
Term
Humoral Immunity
Definition
immunity mediated by antibodies (B cells) that are present in blood, lymph, and other body fluid
-important in the defense against extracellular microbes and microbial toxins
Term
Cell Mediated Immunity
Definition
immunity mediated by T lymphocytes- effect through direct cell-to-cell contact
-important in the defense against intracellular microbes and tumor cells
Term
Innnate vs adaptive receptor differences
Definition
Innate- non-sepcific( respond in a general fashion to foreign stimuli), Recognized different patterns on the surface of antigens, Pattern Recognition Receptors (PRRs) ex. Mannose on bacteria surface or LPS on Gram neg Bacteria
Adaptive- Each B,T Cell has surface receptor BCR, TCR, that are specific for one antigen only and recongize down to single AA differences
-Receptors created by somatic recombination of a gene which allows for great diversity to rearrange genes and give cells varying specificities
Term
Importance of cooperation b/t innate and adaptive immune system
Definition
-innate system is typically the first to respond and then adaptice are second if necessary
-When both are active
-macrophages can also act as APC for T cells
-Antibodies optimize phagocytosis by macrophages and PMN
-Antibodies activate complement system.
-Cytokines produced by cells of the adaptive immune system enhance phagocytic and cytotoxic activity of innate immune cells
Term
Pattern recognition Receptors (PRR)
Definition
Molecular pattern sutilized by cells of the inate immune system to recognize self from non-self (Non-specific)
Term
Receptors for Pathogen Associated Molecular Patterns (PAMPs)
Definition
Mannose receptor, Scavenger receptor, LPS Receptor (gram neg bac)
Term
Opsonin Receptors
Definition
Host derived proteins the coat pathogens,
phagocytes ahve receptors for the proteins bound to the pathogen and can recognize, bind, and phagocytose.
-antibodies Igs, Complement C3b, C-reactive protein CRP.
Term
PhagocyteSpecific Receptors
Definition
can stimulate other immmune responses
Term
Toll like receptors
Definition
-On the surface of phagocytes and expressed primarily by innate cells to recongize various PAMPs that are not found in our own cells such as Flagellin and Viral DSRNA, DNA
Term
Toll like receptors on cell surface look for
Definition
extracellular components such as LPS, bacterial peptidoglycan, flagella
Term
Toll like receptors in endosomal ompartments look for
Definition
intracellular components of microbes such as SSRNA, Viral DNA, anything weird that got in.
Term
What happens when pathogens bind to Toll Like Receptors (TLR)
Definition
stimulates two transcription factors:
NF-kB= promotes inflammation and prepares cellsf or interaction of the adaptive immune system (T-cells), upregulates cell surface expression of costimulatory molecules
IRF-3= promotes production of antiviral interferons to block viral replication and stimulates NK cells
Term
What affects how the cell will respond to cytokines?
Definition
Receptor affinity and/or number
Term
Cytokine Receptor signals
Definition
released to signal cells that will thell them when to exit circulation and tell them where to go (migration)
-act as autocrine or paracrine
-NEVER stored, produced in response to microbes or other antigens
Term
Oxygen dependent vs independent killing
Definition
independent-using various proteolytic enzymes
dependent- Respiratory burst= cellst taking up a lot of o2 to make ROS, RNS; synthesizing NO, superoxide anion and H2O2 that can be used to destroy bacteria.
Term
NK killing procedure
Definition
-NKs only respond to cells not expressing Class 1 MHC molecules
-MHC inhibits killing, w/o it NK cell is activbated and releases cytotoxic substances into the infected cells which leads to apoptosis.
Term
Granular enzymes and function in NK
Definition
Perforin- pore forming protein that inserts into membrane and forms a proe that allows granzymes to pass
Granzymes= activate the caspase pathway that results in apoptosis
Term
Type 1 Interferon Cytokines
give abbreviations
Definition
IFN-a, IFN-b, IFN-gamma, IL-12,
Term
IFN-a, IFN-b
Definition
antiviral cytokines, activate antiviral mechanisms in neighboring cells by inducing an antiviral state in that cell (the cell dies but hopefully the viral replication is stopped)
-Increase class I MCH expressions, stimulate NK activity, and present antigens to CD8 T-cells (cytotoxicT-cells)
Term
IFN Gamma
Definition
Interferon, cytokine.
Naturally released by T cells and NK cells; used as a treatment for CGD patients.
-Enyhances the production of ROS
-Increases the microbial activity of macrophages
Term
IL-12
Definition
Cytokine, secreted by macrophages to activate NK cells- they in turn directly kill infected cells and release IFN gamma to activate macrophages and increase ROI and lysosomal enzymes
Term
Plieotropy
Definition
ability of a cytokine to have a variety of effects based on what it is interacting with
Term
Redundancy
Definition
ability of a cytokine to have a method for chekcs and balances in which multiple cytokines can produce the same effect
Term
Synergy
Definition
the ability of cytokines to work to gether to produce/augment a response
Term
Antagonism
Definition
cytokine ability to work in opposition to eachother, allowing signals to be turned on/off
Term
The four functions of the Complement System
Definition
1. Oposonization= C3b C4b
2. Clearance of immune complexes (C3b binds to immune complexes)
3. Membrane attack lysis (C5b)
4. Inflammation and Chemottraction (C3 and C5)
Term
Complement Opsonization
Definition
C3b an C4b
-proteins coat microorganisms to enhance phagocytosis
-C3b can be protolyticlaly cleaved into iC3b which is an opsonin that can't further amplify the enzyme cascade.
Term
Complement Clearance of immune complexes
Definition
C3b binds to immune complexes, serve as chemoattractant facotrs
Term
Complement Membrane attack lysis
Definition
C5b is the starting point for MAC generation
C5b binds to C6,7,8, and multiple copies of 9. This forms a large pore on the surface of the pathogen allowing water to rush in and lyse the cell
Term
Complement Inflammation and Chemoattraction
Definition
C3a and C5a are chemoattractants and anaphylatoxins
-recruit phagocytes to area of infection and cause release of inflammatory mediators
-"a" cells create concentration gradient that attracts cells to the area.
-Also bind mast cells- degranlation and release of histamine
Term
Formation of C3 Classical Pathway
Definition
C1 binds to antibodies (IgG or IgM) that are bound to pathogen surface
-Conformation shape change activates C1- cleaves C4 into C4a,b
-C4b binds to pathogen surface and C2 binds to C4b
-C1 cleaves C2 into C2a,b (a leaves)
-C4b2b is formed= C3 convertase, cleaves C3 into C3a,b
Term
Formation of C3 Lectin Binding Pathway
Definition
Liver stimulated by cytokines to secrete Mannose-Binding Lectin, which binds to the Mannose on pathogen surface.
-MASP binds to MBL, forms MBL-MASP complex that forms just like C1
-MBL-MASP cleaves C4 to C4a,b
-C4b binds to pathogen surface, C2 binds to C4b
-MBL-MASP cleaves C2 to C2a,b forming C4b2b= Convertase enzyme
Term
Formation of C3 Alternate Pathway
Definition
Spontaneous or pathogen initiated cleavage of C3
-C3b binds to pathogen surface and provides binding site for Factor B, C3bB formed
- Factor D cleaves B into Ba,b
-C3bBb formed= convertase enzyme (stabilized by Properdin)
Term
Describe what can happen to C3 after C3 convertase is formed
Definition
C3 convertase is the convergence of all three pathways
-Some C3b binds to pathogen surface
-Some C3b binds to C3 convertase forming C5 convertase
Term
Explain C5 convertase
Definition
It is C3b bound to C3 convertase
-It cleaves C5 to C5a,b, which serves to aid lysis, opsonization, activate inflammatory response, and clearance of immune complexes.
-C5b is the starting point for MAC-cell lysis
Term
C1 inh
Definition
functions to inactivate the classical and lectin pathway by inhibinting protease activity via C1 and MASP.
-turning off C1 and MASP limits how much C3a and C5a are produced.
=Hereditary Angioedema
Term
Hereditary Angioedema
Definition
C1 and MASP are always active, no C1inh. They continuously generate anaphylatoxins causing excessive swelling most common in the mouth. THey are not able to turn off complement.
Term
CD59 and DAF (CD55)
Definition
These are host regulatory proteins on the cell surface of RBCs.
Term
CD59 and DAF(CD55) defects
Definition
Hemolytic anemia and venous thrombosis
excessive complement activation evident on RBC- generation of MAC complex formation on RBC- RBC lyse- Recurrent intravascular hemolysis.
Term
Immunodeficiency associated with classical protein C1,2,4
Definition
increase the predisposition for developing immune complex diseases- persistence of immune complexes- inflammatory mechanisms continuously activated= tissue damage and dsease such as lupus and SLE
Term
Immunodeficiency associated with C3 deficiencies
Definition
all of the pathways will be deficient- immune complexes diseases, recurrent bacterial infections- defective opsonins and lysis
Term
Immunodeficiency associated with MAC deficiency
Definition
C6 or C9 issues- unable to generate MAC- increased risk of certain bacterial infections- Gonorrhea and meningitis
Term
5 points of acute inflammation
Definition
-Rapid and short lived (minutes to days)
-Dilation
-Inc permeability of vasculature (cells and proteins escape)
-Inc expression of CAMS on endothelium
-influx of PMNs into the tissue (halmark sign)
Term
What inflammatory mediators result from tissue damage?
Definition
Clotting factors and bradykinin
Term
Explain the purpose and synthesis of bradykinin
Definition
Kallikrein converts Kininoge to bradykinin.
-inc vascular permability, vasodilate, promote pain, sm M contraction
Term
Explain events in endothelial damage with the clotting cascade
Definition
endo damage- clotting cascade- thrombin- fibrin degradation- inc vascular permeability, neutrophils, chemotaxis
Term
Explain events in endothelial damage with the fibrinolytic system
Definition
Endothelial damage- fibrinolytic system- plasmin- complement activation- increased vascular permeability, neutrophil, chemotaxis, opsonization, cell lysis.
Term
Anaphylatoxins and #s
Definition
C5a and C3a
-complement proteins that can serve as chemoattractant factors for neutrophils or bind to mast cells- histamine release- increased permeability and dilation
Term
histamine
Definition
released from mast cells and causes dilation and increased permeability
Term
Arachadonic acid metabolites
Definition
Prostaglandins, leukotrienes, thrombozane,
-derived from membrane phospholipids
Supporting users have an ad free experience!