• 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

1/314

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;

314 Cards in this Set

  • Front
  • Back
what mhc would process mhc cells?
mhc 1: considered inside the cell
NK cells are considered what kind of cells
large, granular lymphocytes
what kind of activity do NK cells have?
cytotoxic activity against tumor cells and some virus infected cells
Are NK cells part of the innate or adaptive immune system?
innate: they do not have specific receptors for antigen
ADCC
antibody-dependent cell-mediated cytotoxicity

ex. ab coats antigen and NK cell has a CD16 receptor that interacts with region on AB molecule causing NK-cell cytotoxicity
monocytes are found where?
in the blood
macrophages are found where?
in the tissue
How do monoctyes become macrophages
-they circulate in the blood for about 8 hours, during which time they enlarge; they then migrate into tissue and differentiate into specific tissue macrophages
how can macrophages be activated
-phagocytosis of antigen
-contact with receptors that sense molecules present in microbial pathogens
-cytokines secreted by Th cells
-mediators of the inflammatory response
-
macrophages have what enzymes
hydrolytic
3 big things macrophages do
-hydrolytic enzymes kill microbes
-secrete inflammatory mediators
-increased ability to activate T cells

Activated macrophages also release cytokines
how do Th and macrophages relate to each other
they both help to facilitate each other's activation during the immune response
phagosome
-vessicle after phagocytosis

-if destroying the particle, it will bind with a lysosome to form a phagolysosome and the lysosomes hydrolytic enzymes will digest eh ingested material.....digested material will then be released via exocytosis
opsonin
a molecule that binds to both antigen and phagocyte, theyreby enhancing phagocytosis
3 granulocytes
-basophils
-neutrophils
-eosinophils
neutrophils
-first on site of inflammation
-phagocytic
-highest amount in serum
-motile: can go from blood to tisue
-granules contain hydrolytic enzymes
extravasation definition
-movement of circulating neutrophils into tissues
eosinophils
-motile
-phagocytic
-granules contain hydrolytic enzymes
-useful against parasites
Basophils
-non-phagocytic
-release pharmacologically active substances from their cytoplasmic granules
-allergic response
-contain histamine in granules
-found in blood

Mn: allergic to basil
Mast cells
-tissue form of basophils
Dendritic Cells
-APC
-costimulatory signals
-reactive oxygen species
-cytokines
primary lymphoid organs
-bone marrow
-thymus
medulla
inner component of a tissue formation
cortex
outer compartement of a tissue formation
location of thymus
Above the heart
What happens to the thymus with age?
-with age, the size of the thymus decreases, as does the amount of T cells produced
interstitial fluid
via thin walls in capillaries, plasma seeps out

-the fluid that permeates all tissues and bathes all cells is called interstitial fluid
what happens if interstitial fluid is not returned to the circulatory system
edema
spleen location
high in left abdominal cavity
components of innate immunity
-physical, chemical and cellular barriers

-skin and mucous membranes
-acidity of stomach and specialized soluble molecules that possess antimicrobial activity
-various cells
innate immunity specificty
it is specific for molecular pattern associated with pathogens
self/non self discrimination of innate vs adaptive
innate: perfect because there are no microbe-specific patterns in host

adaptive: very good: occasional failures with self/nonself discrimination
major cells types of innate immunity
Phagocytes: monocytes, macrophages, eosinophils, neutrophils

-NK cells

-dendritic cells
major cells of adaptive immnity
T and B cells
is complement system innate addaptive?
both
-can be activated by ab or my molecules that recognize PAMPS
3 big things complement can do
-opsonization
-lysis by MAC
-inflammation from components it releases
TLR
-detect microbial products and are part of the innate immunity
4 signs of inflammation
-swelling
-redness
-heat
-pain
major role of chemokines
-subgoup of cytokines

-signature activity is their capacity to act as chemoattractants (agents that cause cells to move toward higher concentrations of the agent)
CAMs
cell adhesion molecules

Hold are tissues together and are used by leukocytes to interact with tissue cells
extravasation
when circulating leukocytes enter inflamed tissue or peripheral lymphoid organs, the cells must adhere to andd pass between the endothelial cells lining the walls of blood vessels
do most receptors bind more than one chemokine?
yes
can chemokines bind to more than one receptor?
yes, many can bind to more than one receptor
do leukocytes normally have access to injury area?
no because they are found in the blood vessels
fenstrations
slits between blood vessel epithelium
chemotaxis is...2 parts
1) going out of the BV
2) Going to site in tissue
4 steps of leukocyte extravasation
-inflammation causes cytokines to be released that bind to epithelium and cause several receptors to be expressed on epithelium

1) rolling, mediated by selectins
-cytokines induce expression of adhesion molecules of the selectin family on endothelium wall
-these E and P selectin molecules bind to mucin like cell adhesion molecules on the leukocyte membrane
-process tethers the leukocyte to the endothelial cell....but the shearing force of the circulating blood detaches the cell.....selectin molecules on another endothelial cell again tether the leukocyte: process is repeated so that the cell tumbles end over end .....caled rolling

2)activation by chemoattractant stimulus
-rolling slows cell enough to allow interaction between chemokines
-chemokines on endothelium and chemokine receptors on leukocyte provide a degree of specificty to the recruitment of WBC's to infection site

3) arrest and adhesino, mediated by integrins binding to Ig-family members
-chemokines on endothelium binding to chemokine receptors on leukocytes causes a conformation change and the clustering of integrins> leads to arrest

4)transendothelial migration
-arrest allows for leukocyte to squeeze out of the blood vessel into the tissues: it does this by associating with a variety of receptors
-

see page 332: 13-4
initially what do cytokines do in the early part of an inflammatory response
-cytokines and other mediators act on the local endothelium, inducing expression of adhesion molecules of the selectin family
talk about the molecules involved in T cell extravasation
1) during an inflammatory response, cytokines act on endothelium wall creating expression of adhession molecules for selectin family that is found on leukocytes
-CD34 binds to L-selectin

2) this process causes rolling and the cell slows down
-this causes chemokine found on endothelium to react with leukocyte chemokine receptor....leads to activation

3)Activation leads to conformational changes that cause change in shape and expression of LFA-a that will bind to ICAM-1 on endothelium wall

4)after arrest, cell will bind to various that allow it to pass through endothelial layer
-PECAM-1 binds to PECAM-1
-CD11a/CD18 on leukocyte binds to JAM-1 on endothelium

pg 336
CD34
-Mucin carbohydrate found on endothelium in the initial part of the extravasation of leukocytes

-Bind to Selectins
selectin
-attached to leukocytes
-bind to mucin on endothelial cells
-this binding slows cell down during the rolling phase

correction......involved during early part of rolling......can be found on both leukocyte or endothelial
LFA-1
-an integrin involved in naive T cell extravasation through high-endothelial venule
-binds to ICAM-1
-involved in arrest/adhesion after cells has come into contact with its chemokine
ICAM-1
-Ig superfamily CAM's
-binds to LFA-1
-involved in arrest of cells
talk about selectin vs integrin binding
selectin: weak
integrin: stronger
what does E-selectin bind to
-E selectin is found on the endothelium....binds to mucin

***Remember selectin can be found on both leukocytes and the membrane...just as can mucin

-It binds to s-Lex expressed on RBC and WBC
s-Lex
-found on RBC and WBC
-Binds to E-selectin
E-selectin
on endothelium
binds to s-Lex
chemoattractants
1)formylated peptides
-fMet-leu-phe: bacterial proteins start with formylated peptides

2)complement anaphylotoxins: C5a, C3a

3)Chemokines
-CC chemokines : aa next to eact other
-CXC chemokines: spread out by 1 aa
MCP-1
-produced by
-receptors
-cells attracted
-major effects
-a CC chemokine

-monocytes, macrophages, fibroblasts, keratinocytes

-CCR2B

-attracted: monocytes, NK and T cells, Basophils, dendritic cells

-: effect: activates macrophages, basophil histamine release
Eotaxin
-produced by
-receptors
-cells attracted
-major effects
CC chemokine

-produced by: endothelium, monocytes, epithelium, T cells

-Receptors: CCR3

Cells attracted: Eosinophils, Monocytes, T cells

-effect: role in allergy
IL-8
-produced by
-receptors
-cells attracted
-major effects
CXC chemokine

produced by: monocytes, macrophages, fibroblasts, keratinocytes, endothelial cells

receptor: CXCR1, CXCR2

cells attracted: neutrophils, Naive T cells

Effect: mobilizes, activates and degranulates neutropils, angiogenesis, acts as a neutrophil chemoattractant
IP-10
-produced by
-receptors
-cells attracted
-major effects
CXC chemokine

-produced by: keratinocytes, monocytes, T cells, fibroblasts, endohelium

receptor: CXCR3

Cells attracted: Resting T cells, NK cells, Monocytes

Major effect: immunostimulant, antiangiogenic, promotes TH1 immunity
talk about co-receptor for HIV
-co-receptor is chemokine reeptor R4 or 5
-when chemokine binds to receptor, HIV can't bind

also......HIV uses CD4 and chemokine receptor
does ligand always bind to same receptor?
no
CCR2 binds to what
CCL2 (MCP1): is a CC subgroup of chemokines
draw a simplified signal transduction pway for chemokine receptor
pg 331
CCR7
-if found on a cell, it can be assumed that the cell is a memory T-cell
chemokine receptors as activation markers on different cell groups:

draw groups
page 331
chemokine receptors are mediators of what?
cell movement
how do lymphocytes get back into blood?
via the thoracic duct
venules are what?
-sticky, cuboidal high shape ( cells are normally flat)
-lymphocytes adhere to high endothelial venules: mediated by selectins and integrins
-account for small area of endothelium
-85% of bound cells adhere to these even though they only take up 1-2% of area
-HEV's express several adhesion molecules

-see page 335
Draw scheme of lymphocyte recirculation routes:
page 334
define the following
selectin
mucin
integrin
ICAMs
selectin: membrane glycoproteins has an extracellular lectin like domain that enables the molecules to bind to specific carbohydrate groups
-has L, E and P selectin
-circulating leukocytes express L-selectin
-C and P selectin are expressed o vascular endothelial cells during an inflammatory response

Mucin
-serine and threonine rich proteins
-bind to selectin
-found on endothelium and leukocytes

integrins:
-heterodimeric with alpha and beta chain
-bind to ICAMs
-found on leukocyte

ICAMS
-Ig domain-
-bind to integrins
what is a superfamily
largest family with shared structure

genes and proteins are similarly organized
members of the Ig superfamily
-IgG
TCR
MHC
CD4 and CD8
ICAM
B7
Etc
How does endothelium become more sticky?
-By putting P-selectin on surface
draw 3 selectin molecules and describe their function
lecture 14: 7c
draw 3 leukocyte integrins
lecute 14: 8a
draw the breakdown of membrane phospholipid in the generation of inflammatory mediators
pg 339

-many drugs inhibit cycloosygenase pathways and lipoxygenase pathway

-prostaglandin and leukotriene A mediate inflammation
Draw big overall picture of inflammation from end of chapter 13
pg 341
is inflammation a positive or negative feedback loop?
positive
4 functions of cytokines
can have
-pleiotropic
-redundant
-synergistic
-antagonistic function
how can cytokines and their receptors be grouped?
-based on common structures, subunits, and signaling machinery
major signaling mechanism for many cytokines
JAK/STAT system
chemokines use what receptors to signal in what way?
-they use serpentine (7-transmembrane) receptors to signal via trimeric G-proteins
cytokines and chemokines allow for what?
allow for the fine shaping and fine-tuning of an immune response as it progresses
are cytokines soluble?
yes....they are small proteins
cytokines part of innate or adaptive?
both
cytokines mediators of what?
-innate
-adaptive
-mechanism by which leukocytes communicate with one another
-5 effects that cytokines can have
-stimulation
-inhibition
-differentiation
-cell death
-chemoattract
show autocrine, paracrine ad endocrine action of cytokines
pg 303
scheme with 5 properties of cytokines

pleiotropy....etc
pg 304
cytokines of innate immunity:6
cytokines of adaptive immunity:5

Secreted by?
Targets and effects?
pg 307
local infection with gram negative bacteria vs systemic infection with gram negative bacteria
local
-macrophage activated to secrete tnf alpha in the tissue
-increased release of plasma proteins into tissue, increased phagocyte and lymphocyte migration into tissue, increased adhesion of platelet to blood vessel wall
-phagocytosis of bacteria: plasma and cells drain to local lymph nodes
-removal of infection and adaptive immunity

systemic
-macrophage activated in the liver or spleen secrete TNF alpha into the blood stream
-systemic edema caused decreased blood volume, hypoproteinemia, neutropenia, followed by neutrophillia....decreased blood volume causes collapse of blood vessel
-disseminated intravessicular coagulation leading to wasting and multiple organ failure
-death

Sepsis causes abnromal Temps, WBC counts, respiratory rate, followed by capillary leakage, tissue injury and lethal organ failure
virus infected host scheme
lecture 15: 4a
cytokine secretion and principal functions of mouse TH1 and TH2 subsets
pg 315
functions of cytokines in acquired immunity

-cytotoxic
-TH1
-TH2

Draw what molecules would be released
lecture 15: 4c
IL4
IL5
IFN gamma
TGF beta

Role of these cytokines in regulating Ig isotype expression
lecture 15: 5a
5 cyokine receptors and a few members of each family
pg 308
draw and describe class 1 cytokine subfamilies and explain importance
pg 309
cytokine receptor subfamilies have what?
shared signaling subunits
draw IL-2R
pg 311

-signal transduction is mediated by the beta and gamma chains, but all three chains are required for high-affinity binding of IL-2

-beta and gamma are considered a member of class 1, but alpha is not
-alpha only involved in affinity for antigen

-gamma chain is constitutivly expressed but beta and gamma are more expressed when exposed to antigen

-this ensures that only Ag activated T cells will express the high affinity IL-2 receptor
talk about IL-2Ralpha
expressed only by activated T cells: beause of this, it is sometimes referred to as the TAC (T-cell activation) antigen or CD25;
draw pathway that class 1 and class 2 cytokine receptor signl by....
pg 312
JAK/STATs Used by Key cytokines

-IFN-gamma
-IL-2
-IL-4
-IL-12
page 312
what structure is predicted to be similar to chemokine receptor?
retinal (when rhodopsin is bound to it)
what kind of receptor are chemokines
G proteins.....signal by coupling with trimeric GTP-binding proteins
talk about IFN-g
macrophage and NK cell activation

-associated with activation of TH1 cells

-cell mediated immunity (T cell activation)

-Inflammation

-viruses, tuberculosis, Leishmania

-Tissue immunity
talk about IL-4
antibody production

-humoral immunity: Ab production

-anti-inflammatory

-worms, extra cellular-Bacteria, fungi, toxins, et

-surface immunity
processing of an antigen by APC and presentation to T cell leads to what?
-T cell activation and cytokine production
scheme of TH1 activation, TH2, and TH17
-ag internalized and presented by APC

-TH1 activated by release of IL-12, p40/p35>>>>>>>leads to CMI and IFN-g

-APC automatically activates Th2 with presentation of Ag>>>>>>>leads to IL4

-APC activates TH17 via IL-23 and p40/p19>>>>>>>>leads to IL-17

see lecture16: 2a
activation of Th17 leads to what?
-PMN-recruitment
-inflammation
-IL-17 creation
cd40
a co-stimulatory molecule found on APC....must bind to CD40L found on T cell to become active

-CD40L is found on CD4 cells and binds to CD40 on APC cells
fas ligand
-found on CTL

-binding of fas to its receptor leads to apoptosis
TH1 activates what big thing
-macrophasge

-TH1 may express Fas ligand and cause macrophage to go through apoptosis
TH2 activates what big thing
-B cells
IL-13
secreted by TH-2 cells

important mediator of allergic inflammation and disease.
scheme of cytokine-mediated generation and cross-regulation of Th subsets
pg 316
vaccinia virus
-member of pox virus family
-vaccine to erradicate smallpox
*different immune response to different pathogens: CTL, TH1 and TH2 for following

-typical pathogens
-locations
-antigen recognition
-effector action
cytotoxic CD8 T cell
-Typical Pathogens: Vaccinia virus, influenza virus, Rabies Virus, Listeria
-Location: Cytosol
-Antigen Recognition: Peptide MHC class 1 on infected cell
-Effector action: killing of infected cell

TH1
-pathogens: Mycobacterium tuberculosis, Mycobacterium leprae, Leishmania donovani, pneumocystis carinii
---Location: macrophage vessicle
---Ag recognition: peptide MHC class 2 on infected macrophage
---Effector action: Activation of infected macrophage

TH2
-pathogens: Clostridium tetani, Staphylococcus aureus, streptococcus pneumonia, polio virus, pneumocystis carinii
---location: extracellular fluid
---Ag recognition: peptide/mhc class 2 on Ag specific B cell
---Effector: activation of specific B cell to make Ab
leishmania donovani
-infects macrophage vesicles
-cleared by Th1 response
-type of trypanosome protozoa
-Causes Leishmaniasis from a sand flye bite
-Sx of fever and skin sore from where bite occures

Mn: picture a gigantic leesh on a flye infecting macrophages with a protozoa
listeria
-gram positive bacteria
-infects cytosol of cells
-cleared by CTL
-Listeriosis symptoms include vomiting, nausea, stomach cramps, diarrhea, severe headache, constipation, persistent fever, stiff neck, loss of balance and convulsions

Mn: listeria gets into your mouth cells
Rabies Virus
-found in cytosol of cells
-cleared by CTL response
-Sx: encephalitis (inflammation of the brain)

In the beginning stages of rabies, the symptoms are malaise, headache, and fever, while in later stages it includes acute pain, violent movements, uncontrolled excitements, depressions, and the inability to swallow water (hence the name hydrophobia). In the final stages, the patient begins to have periods of mania and lethargy, and coma. Death generally occurs due to respiratory insufficiency
influenza virus
-infects cytosol of cells
-cleared by CTL response
Pneumocystis carinii
-pneumonia caused by yeast like fungus

-same as pneumocystis pneumonia

-cleared by TH1 or TH2 response

-symptoms of PCP include fever, non-productive cough, shortness of breath (especially on exertion), weight loss and night sweats. There is usually not a large amount of sputum with PCP unless the patient has an additional bacterial infection.
clostridium tetani
Clostridium tetani is a rod-shaped, anaerobic bacterium of the genus Clostridium. Like other Clostridium species, it is Gram-positive

-cleared by TH2 response

Mn: if you step on a nail, for positive it will hurt
staphylococcus pneumonia
-gram positive

-cleared by Th2 response

-
streptcoccus pneumonia
-gram positive bacteria

-cleared by Th2 response
polio virus
-spread via fecal oral route
-infects GI tract
-can cause CNS damage leading to paralysis

-cleared by TH2 response

Mn: goes in hole number 2
scheme of worms vs viruses and what becomes activated
lecture 16: 3b

Viruses and some bacteria induce IL-12 secretion by dendritic cells that can activate NK cells to produce IFN-g..........
Natitive CD$ T cells activated in the presence IL-12 and IFN-g are committed to differentiate into Th1 cells


Other pathogens...worms...do not induce IL-12 expression by dendritic cells but cause the synthesis and secrtion of IL-4>>>>>>>>>>>>>>Natitive CD4 T cells activated in the presence of IL-4 are committed to differentiate into TH2 cells
complete following for
IL-2
interferon gamma
lymphotoxin
IL-4
IL-5
IL-10

T-cell source,
Effects on following:
-b cell
-t cell
-macrophages
-hematopoitic cells
-other somatic cells
-effect of gene knockout
lecture16: 3c, 4a
B7
-signal 1 is generated by interaction of Ag peptide with the TCR-CD3 complex

-signal 2 is from interaction between CD28 on T cell and members of the B7 family on the APC.....

-ligand for B7 can also be CTLA-4 which is an inhibitor ligand>>>>down regulation of T cells
Th1 response and macrophage activation scheme and what would occur
-lecture16: 4b,c
DTH=
DTH=CMI
DTH response scheme
lecture 16: 5a
tuberculosis is what response
Th1
explain Tb granuloma
-cd-4+ TH1 cells are activated within 2-6 weeks after infection, inducing the infilitration of large numbers of activated macrophages

-These cells wall off the organism inside a granulomatous lesion called a tubercle

-tubercle consists of a few small lymphocytes and a compact collection of activated macrophages, which sometimes differentiate into epithelioid cells or multinucleated giant cells

pg 459
-lecture 16: 5b
lytic cycle results in what?
destruction of host cell
mycobacterium leprae tidbits
there are 2 polar forms: several intermediates exist

tuberculoid leprosy:
-organisms present at low to undetectable levels
-low infectivity
-granulomas and local inflammation peripheral nerve damage
-normal serum immunoglobulin levles
-normal T-cell response specific for M. Leprae ag
-cell mediated forms granulomas
-slow progression
-Th1 response...with delayted type sypersensitivity: IL-2, IFN-g, TNF-B

Lepromatous leprosy
-organisms show florid growth in macrophages
-High infectivity
-disseminated infection: bone, cartilage, and diffuse nerve damage
-hypergammaglobulinemia
-low or absent T-cell responseiveness
-no response to M. leprae antigens
-Th2 response: IL-4, IL-5, IL-10
leishmania lives where?
it is an intracellular parasite that lives in macrophages phagosomes
talk about leishmania
-lives in macrophage phagosomes

-protozoan parasiite caused by Leishmania major

-different individuals can have different Sx due to immune response

-resistant individuals develop a TH1 response and clear the organism via IFN-g; non-resistant individuals develop a TH-2 response and do not clear the organism
talk about BALB/c and leishmania
BALB/c mice are highly susceptible to Leishmania and frequently succumb to infection.....these mice mount a TH2 response ....they produce high levels of IL-4 and no IFN-g

-applying IL-12 to BULB C mouse makes response almost as effective as normal cells
what is an important source of IFN-g?
-cytotoxic CD8 t cells
draw TH2 respone
lecture 16: 7b
what response is involved in allergy?
TH2..leads to IgE production
schistosomiasis
TH2 response=worm infection
Eosinophils
parasitic worm

-invades skin, travels to liver, reproduces

-reaches maturity at 6 to 8 weeks and starts to produce eggs: eggs normally get taken from body via digestive tract, but many become trapped in the mesenteric veins, or will be washed back into the liver, where they will become lodged.

-rapped eggs mature normally, secreting antigens that elicit a vigorous immune response. The eggs themselves do not damage the body. Rather it is the cellular infiltration resultant from the immune response that causes the pathology classically associated with schistosomiasis.

-abdominal pain, cough, diarrhea,

-worse: hepatosplenic enlargement and dermatitis
S. Japanicum
type of schistomiasis
worm infection=
Th2 Response
dissociation constant of receptor
lecture 16: 9b
NK cells fall under what group of cells
lymphocytes
what are the effector cells of CD8+ cells
CTL
Do Naive CD8+ T cells and CTL have different requirements for activation?
yes, particularly regarding APC and costimulation
how do CTL kill infected and tumor cells?
by inducing apoptosis
main effector molecules of CTL
perforin, granzymes, granulysin......all of which are found together in lytic granules........and FasL (a surface-bound TNF family member)
perforin
one of main effector molecules of CTL along with granzymes, granulysin......all of which are found together in lytic granules.......and FasL......a surface-bound TNF family member
Think what, when you see Fas
TNF
what happens when receptor is discovered first?
-when receptor is discovered first, its ligand ends up getting an L after it
Naive CD8+ T cells do not have what? and have what?
do not have cytotoxic activity

-have a stringent requirement for costimulation
upon activation, naive CD8+ T cells can do what?
differentiate into CTL: production of lytic granules, acquisition of killing activity, IFN-g secretion
CTL have minimal or no requirement for....
costimulation: most target cells lack B7 molecules
co-stimulatory molecules are only presented on what?
professional APC
draw scheme of licensing

pg 354
lisensing basically primes the dendritic cell to activate cd8 cells

1)initial licencing occurs when dendritic cells interact with TH1 cells or are activated by TLR when the Ag binds to it

2)dendritic cell now shows MHC class 1 and has costimulatory molecules B7

3)Activatin of CTL-P occurs and it now bears the IL-2 receptor

4)IL-2 is released from TH1 cells and it binds to CTL-P cell causing it to become cytotoxic
TLR function
found in innate immunity
talk about memory CTL's
pg 355

antigen activated memory CTL-Ps appear to secrete sufficient IL-2 to stimulate their own proliferation and differentiation into effector CTLs.....they also may not require the CD28-B7 costimulatory signal for activation
what are the CTL's main weapon?
lytic granules....they are like smart bombs
lytic granules are what?
modified lysosomes that hold effector molecules in conditions that prevent activity
what causes effector molecules to be released from lytic granules?
calcium flux induced by TCR signaling causes release of granules toward target cell
Once released, what do lytic granule contents do?
they become activated and induce programmed cell death in target cell
essentially what do proteases do?
chew up proteins to degrade them
apoptosis in the immune system

-apoptosis=

-considered what kind of cell death?

-induced by what?

-activation of what causes death?
-apoptosis=programmed cell death

compartmentalized cell death: cells die without spilling contents, and express signals for rapid phagocytosis....phosphatidylserine

can be induced by developmental cues, cellular damage, or signals from cytotoxic cells (CTL, NK cells)

Activation of cysteine proteases, the caspases, is signal to die and also initiates cell death process
Features of Apoptosis....5 parts
-nuclear condensation

-membrane blebbing: membranes pinch off as small fragment>looks like membrane is boiling away

DNA fragmentation into 200bp pieces....DNA is wrapped around histone and is degraded by proteases at 200bp intervals

-degradation of cellular components

-Exposure of phosphatidylserine on outer membrane leaflet: it usually only on the inner leaflet
is there a lot of inflammation during apoptosis?
no because you are not releasing fragments into the environment
a single differentiated CTL can do what?
can kill many target cells, and does not require costimulation/Th cell help
draw Apoptosis scheme and CTL recycling
pg 356
proteins in lytic granules of cytotoxic T cells and Actions on target cells
perforin: polymerizes to form a pore in target membrane........forms large pore, allowing ions and water to flow into cell-osmotic stress induces apoptosis, but is inefficient

Granzymes: serine proteases, which activate apoptosis once in the cytoplasm of the target cell.......enter cell through perforin pore and induce apoptosis by activating caspases, initiating caspase signaling cascade

Granulysin: induces apoptosis


FasL: not in granules....but a mechanism for how CTL's kill
-membrane-bound TNF family member, binds receptor (Fas) on target cell, activating caspase signaling cascade
draw CTL-mediated pore formation in target cell membrane
pg 358
draw the two pathways of target-cell apoptosis stimulated by CTL's
pg 360

-both pathways can activate procaspase8

-UV radiation causes mitochondria to become leaky>cytochrome c released and binds to procaspase-9

-caspase 3 activation very important
talk about specificity of apoptosis
-t cell recognizes infected cell
-infected cell is programmed for death
-neighboring uninfected cells are not killed
what has allowed mice to become a powerful genetic system?
-the ability to directly alter the genomes of mice
what is a transgenic mouse
-(Tg)

A mouse with an additional gene introduced into its genome
-the gene may be wild-type (normal), mutant, or even from another species altogether
-the gene is usually inserted randomly, and often as multiple tandem copies

......ex. could introduce green fluorescent gene

.....many copies allows high expression levels
Knock out mouse:
a mouse in which a gene has been specifically inactivated by targeted deletion

---this made mice important....can not do this with fruit flyes or C. elegans
inducible transgenic
can be turned off/on at will

ex. via a drug
conditional/tissue-specific knock-outs
only in a tissue; only in a condition

ex: want to affect liver, but not heart
knock in
gene inserted at specific location.......in a transgenic mouse it is inserted at a random location
what made mice important
knock out mice
draw basic scheme for transgenic mouse
-563

-gene insertion is random
Uses and caveats of Tg mice
uses:
-can study the function of any gene by overexpression; some by dominant negative (mutant inhibits function of normal gene)

Can use RNAi technology.......transgene encodes for inhibitory RNA.... to knock down gene>>>>>>this decreases inhibition

Can use tissue-specific promoters to target gene expression:

Can express reporter genes (GFP, B-gal) to visualize protein expression patterns>>>>specific location

Note: transgene is inherited as any dominant Mendelian trait
dominant negative
in Tg mice, mutant inhibits function of normal gene
transgene is inherited as .....
inherited as any dominant Mendelian trait
Caveats of Tg Mice
Lecture 17: 7c
2 schemes for knock-out mouse formation
pg 565 and 566
uses and caveats of KO Mice
lecture 17: 8c
talk about the 2 cool special cases of mice with: inducible transgenes and knock-in mice
lecture 17: 9a and b
draw the time course of viral infection with NK cells as the focus
page 361

-activity stimulated by IFN-alpha and IFN-beta

-NK cells produce IFN-g that that activates macrophages>>>phagocytosis>>>>>macrophages also release IL-12>>>>>activates Th1 subset of cells
signal transduction through TLR's-Type 1 IFN
lecture 18: 2a

page 70

TLR uses pway independent of MyD88
explain how double stranded viral RNAs can be recognized
lecture 18:2c

pg 63
draw flow diagram of a virus infected host cell

lecture 18:3a
18: 3a
draw pway for induction of antiviral activity by IFN-a or b
pg 450
do NK cells express Ag specific receptors?
no
HLA=
HLA= MHC

human leukocyte antigen
2 schemes of inhibitory receptor on NK cells
pg 363

lecture18: 3c and 4a
what does KIR stand for?
killer-cell immunoglobulin-like receptors......found on NK Cells....binds to MHC class 1
what cells have ITIMS?
lecture 18: 4b

immunoreceptor tyrosin-based inhibitory motif
sequence for ITIM and ITAM
lecture 18: 4c
ITIMS do what (easy answer)
antagonize ITAMS
other than antigen receptors, what other receptors associate with ITAM-containing chains to deliver activating signals
lecture 18: 5a

*remember tyrosines become phosphorylated and form docking sites for other proteins ....leads to signal
Activating and Inhibitory Receptors.....picture with all of the receptors

other activating receptors
lecture 18: 5b

activating receptors: NKG2D, NKp30, NKp44, 2B4, CD16

other ligands: ULPB in humans and Rae-1 in mice
NKG2D
an important activating receptor that binds MIC-A and MIC-B.....these ligands are like MHC
other activating receptors
NKp30
NKp44
ligands are induced by.....
stress
adaptors can determine receptor status....example?
CD94:NKG2a------inhibitory signals

CD94:NKG2c-----activating signals
NK cell receptors mediating activities fall into 2 structural families
the lectin-like receptors and the killer-cell immunoglobuline-like receptors
a deficiency of activating events to NK cells can lead to what?
-viral infection
-cancer
-accumulation of damaged, non-functional cells
genes encoding NK receptors
-lecture 8: pg 6a

stands for leukocyte receptor complex
how many KIR genes are there in one organism
9-14
Talk about ligands for NKG2D
ligands for NKG2D are MHC-like molecules, MIC-A, MIC-B, or RAET1 family members, whose expression is induced by cellular stress

-RAET1 incudes ULBP's
Are NK cells always cytotoxic?
-yes, and they always have large granules in their cytoplasm
NK-cell cytotoxic pathway
-page 360

-similar to CTL (same in the picture)
why were NK cells named as they are?
-they are non-specific in their cytotoxicity
percent of NK cells of circulating lymphocytes
5-10%
What activates macrophages
IFN-g
NK cells are considered to be apart of innate or adaptive immunity
both
with regards to NK cell, viral infection leads to what?
decreased MHC class 1 expression on cells and induces NK cell killing
Talk about tumor cells activating NK cell
tumor cells have decreased MHC class 1, but increased stress-related activating NK cell ligands
talk about ADCC

has 2 drawings
lecture 18: 7c and 8a

pg 366

antibody-dependent cell-mediated cytotoxicity
NK cells are an important source of ....
innate IFN-g

IFN-g prevents Th2 class and forms TH1 class.....TH1 class will then form more IFN-g and NK-cells will cease
NKT cell
-third type of cell identified with characteristics common to both the CTL and Nk cell

-hybrid

-has TCR

-do not recognize MHC-peptide complex...but a glycolipid presented

*******important: releases IL-r stimulating TH2 cell lineage, and preventing TH1 lineage from forming and releasing IFN-g
normal respiratory rate
20/min
the first line of defense against viral infections
NK cells
what leads to immunodeficiencies?
defects in function of immune system components
what are primary immunodeficiencies due to?
mutations in immunologically important genes
what are secondary immunodeficiences caused by?
non-genetic factors
Defects in each component (complement, B cells, T cells, phagocytic cells) will do what?
lead to specific deficiencies, based on the specific functions of the defective piece
SCID stands for what?
severe combined immunodeficiency
In SCID, both.........
although.....
bot B and T cell function is impaired, although the primary deficiency may just be in the CD4+ cells
What does treatment of primary immunodeficiences require?
either bone marrow transplant or replacement/repair of the mutated gene (gene therapy)
primary immunodeficiency:
A defect in immune responses as a result of mutation in one or more immunologically important genes: inherited immunodeficiency
inherited immunodeficiency considered what?
primary immunodeficiency
Secondary immunodeficiency
impaired immune response due to extrinsic factors: acquired immunodeficiency
acquired immunodeficiency
same as secondary immunodeficiency....impaired immune response due to extrinsic factors
immunosuppression
intentional induction of immunodeficiency
3 parts of detecting immunodeficiency's
-Generally found during childhood-patients present with chronic infection, often of the same or similar type

-nature of recurrent infection gives clues as to type of immunodeficiency

-Test implicated components of immune system for presence and or function
Types of immnodeficiency (5 types)
lecture 19: 2b
flow chart of immunological defects, where they effect, and what the cause and impaired function is
-pg 494 and 495
What is complement deficiency caused by?
-mutation in gene for specific complement component
What do most complement deficiencies effect?
most affect complement opsonin function---this leads to susceptibility to pyogenic (pus-causing) extracellular bacteria, such as Streptococcus spp. and Staphyloccus spp. (complement required for phagocytosis
pyogenic
pus causing
defects in MAC component lead to what?
C5-C9 lead to selectie susceptibility to Neisseria spp. infections (importance of complement-mediated killing of infected cells)
draw flow chart of classical, lectin, and alternative pathway......what it would cause, specific components involved
lecture 19:3a

-immune-complex: when Ab and complement proteins bind to Ag of invader...these are then cleared by the spleen:......if not cleared they can become trapped in the kidney, lungs, skin or other tissue and lead to disease
B cell/ antibody deficiences generally are in response to what?
extracellular bacteria and viruses
Bruton's X-linked agammaglobulinemia
-Btk mutation

-Block in B cell development, no peripheral B cells, no serum Ig.
Hyper-IgM syndromes
defects in B cell activation and class switching-lots of IgM

littles or no IgA, IgG or IgE

Caused by CD40L mutation (also some T and macrophage defects)

autosomal hyper-IgM caused by mutation in AID (activation-induced cytosine deaminase)......an enzyme involved in class switching and somatic hypermutation
selective immunoglobulin deficiencies
-selective IgA deficiency: unknown cause

-common variable immunodeficiency: IgM, IgG and IgA.....also called Acquired hypogammaglobulinemia

-map to unkown genes in MHC locus
T-cell deficienceis often lead to what?
-SCID.......increased susceptibility to all classes of pathogen
X-linked scid
-gamma constant chain defect

early block in T cell development, so no peripheral T cells
Bare lymphocyte syndromes
-lead to loss of MHC molecule expression

Defects in TAP genes prevent MHC class 1 protein surface expression so no CD8+ T cells

surprisingly mild immunodeficiency..... respiratory and skin infections

---Defects in TF's controlling class 2 gene expression (CIITA, RFXANK, RFX5, RFXAP) block CD4+ T cell development- result in SCID
DiGeorge's Syndrome
(TBX1 deletion)....single-copy deletino of transcription factor T-Box 1, leads to defect in thymic epithelium development (similar to nude mouse).....haploinsufficiency
haploinsufficiency individuals can be said to be what?
-have Digeorge's Syndrome
name some B cell/ antibody deficienceis
bruton's X-linked agammaglobulinemia

hyper-IgM syndromes

selective Ig deficiencies: selective IgA deficiency, common variable immunodeficiency
name some T cell deficiences
X-linked SCID

Bare Lymphocyte Syndrome

DiGeorge's Syndrome

Defects in T cell signaling molecules (CD3 components, ZAP-70)

defects in cytokine expression, cytokine receptors, or cytokine signaling molecules (esp. IL-7)
CD3
coreceptor for TCR on T cells
ZAP-70
binds to phosphorylated CD3 leading to signal transduction pathway
B/T cell deficiencies.....name some
some forms of SCID result in lack of both B and T cells, due to mutations in genes required for development of both

ADA and PNP defects

RAG1/RAG2 defects

Autosomal scid

Ataxia telangiectasia
ADA and PNP defects
ADA (adenosine deaminase) and PNP (purine nucleotide phosphorylase) defects affect purine degradation- no B or T cells. The most common autosomal SCIDs

B/T cell deficiencies
Most common autosomal SCIDs?
ADA and PNP defects
RAG1/RAG2 defects
B/T cell deficiences

recombination-activating genes

proteins act synergistically and are required to mediate V-(D)-J joining
Autosomal Scid
B/T cell deficiencies

(DNA repair enzyme)- defects in DNA repair enzymes lead to unrepaired double strand breaks during TCR and BCR locus rearrangement- B and T cell precursors die
Ataxia telangiectasia
(ATM defect) protein involved in signaling presence of DNA double strand breaks; failure to induce DNA repair machinery during T and B cell development
phagocyte deficiencies lead to what
-phagocytic cells are crucial for immune response to bacteria and defects lead to severe susceptibility to bacterial infection
types of phagocyte deficiencies
-Severe congenital neutropenia

Cyclic neutropenia

leukocyte adhesion deficiency syndrome

defects in intracellular killing mechanisms (chronic granulomatous disease, G6PD deficiency, Myeloperoxidase deficiency, Chediak-Higashi syndrome)
Chediak Higashi syndrome
caused by defect in lysosome-phagosome fusion

phagocyte deficiencies
chronic granulomatous disease
phagocyte deficiencies

cells have trouble forming the reactive oxygen species

caused by defects in NADPH oxidase system (failure to make superoxde radical)
G6PD deficiency
-X linked recessive phagocyte defect

-enzyme that protects RBC's

-leads to bacterial infection
can a T cell defect affect both humoral and cell mediated parts of the immune system?
-yes because of its central roll in the immune system
pathogenic agents associated with humoral deficiency vs cell-mediated deficiency
humoral: extra-cellular bacteria

cell-mediated: viral, protozoan, fungal.....all intra-cellular pathogens
definition of SCID
stems from defects in lymphoid development that affect T cells, either alone or in combination with B cells and NK cells

-T cell are always gone

-If T and B and NK cells are gone, it becomes more serious
Myeloperoxidase deficiency
Myeloperoxidase deficiency is a common genetic disorder featuring deficiency, either in quantity or function, of myeloperoxidase, an enzyme found in certain phagocytic immune cells, especially polymorphonuclear leukocytes.

phagocyte deficiency

-decreased ability to kill bacteria
neutropenis
reduced number of neutrophils

Severe congenital neutropenia: can be dominant or recessive....Neutrophil counts are persistently less than 0.2E9/liter (less than 10% of normal).....fatal without bone marrow transplant

Cyclic neutropenia (neutrophil elastase mutation)- neutrophil numbers cycle between normal to very low/ none over 21 day period. Other hematopoitic cells have similar, but less severe, fluctuations....due to changing fluctuations of cell production via the bone marrow
Leukocyte adhesion deficiency syndrome
defect in integrin common B2 subunit CD18...are required to facilitate cellular interaction

effects migration of leukocytes to area of infection

causes gram-1 and gram-+ infections and fungi
integrin family
functions as adhesion molecules and are required to facilitate cellular interaction

LFA-1, Mac-1 are examples

have common B chain CD18
name the associated infectious or other diseases of the following syndromes

LAD

Chronic granulomatous disease

G6PD deficiency

Myeloperoxidase deficiency

Chediak-Higashi syndronme
lecture 19...4c
other primary immunodeficiences
cytokine defects- mutations in IL-12, IL-12R, or IFN-γR
lead to susceptibility to otherwise non-pathogenic
intracellular bacteria (esp Mycobacterium spp. and
Salmonella spp.)

X-linked lymphoproliferative syndrome (SAP defect) -
defective control of Epstein-Barr virus (cause of
mononucleosis) leads to B cell hyperproliferation, liver
necrosis, and death. SAP is involved in regulating IFN-γ
production by T cells in response to EBV infection.
X-linked lymphoproliferative syndrome
X-linked lymphoproliferative syndrome (SAP defect) -
defective control of Epstein-Barr virus (cause of
mononucleosis) leads to B cell hyperproliferation, liver
necrosis, and death. SAP is involved in regulating IFN-γ
production by T cells in response to EBV infection.
treatments and problems for immunodeficiencies
Bone-marrow transplantation (BMT) - requires at least one
MHC allele in common between graft and patient.

problem......Potential problems with bone marrow transplantation:
GVHD (graft vs. host disease) - mature T cells in the bone
marrow graft attack the patient’s tissues (due to
alloreactivity)
HVGD (host vs. graft disease) - graft is rejected by the
patient’s mature T cells

Gene therapy - replace defective gene in patient’s own
lymphocytes or hematopoietic stem cells. Limited success
with ADA gene therapy using mature lymphocytes or cord
blood (HSCs). Better success with X-linked scid and
retroviral transduction of bone marrow, but leukemia risk
scheme of a bone marrow graft
lecture 19: 5c
talk about gene therapy
Gene therapy - replace defective gene in patient’s own
lymphocytes or hematopoietic stem cells. Limited success
with ADA gene therapy using mature lymphocytes or cord
blood (HSCs). Better success with X-linked scid and
retroviral transduction of bone marrow, but leukemia risk
talk about Secondary immunodeficiencies
Malnutrition leads to general immunosuppression, often
associated with lethal measles and tuberculosis. May be
links to endocrine system (steroids, leptin)

Diabetes is associated with immunosuppression

Hematopoietic tumors (leukemias and lymphomas) can
cause immunodeficiency due to neutrophil dysfunction or
destruction of lymphoid tissues

Medical interventions - chemotherapies/radiation therapy
(kill bone marrow-derived cells); implanted medical
devices lack innate immune responses, and serve as sites of
“immune privilege” for opportunistic infections

AIDS - depletion of CD4+ T cells by HIV infection
summary of immunodeficiencies

......immune system defect with specificity and susceptible to what organism?
lecture 19....6c
4 types of hyper sensitivity responses
372
general mechanism for type 1 hypersensitivity
373

lecture 20: 3a, 3b, 3c
common allergens associated with type 1 hypersensitivity
374
what about house dust mite
fecal pellets contain Der p1 protein which people are allergic to
talk about the FcERI high affinity receptor
376
signal transduction pathway of mast-cell activation and degranulation
-378
kinetis of major biochemical events that follow cross-linkage of bound IgE on cultured human basophils with igE fragments
379
principal mediators involved in type 1 hypersensitivity
380
biological effects of activating mast cells and eosinophils
lecture 20: 5b
testing for an allergan
-Wheal and Flare Reaction

-When people undergo skin testing for allergies, allergens are rubbed onto their skin and then the skin is pricked to see if a wheal and flare reaction appears. If no reaction is observed, allergens may also be injected under the skin. Injection triggers a more intense reaction, making a wheal and flare clearly visible even if someone is only mildly allergic.

A wheal and flare reaction is a skin reaction which occurs in response to exposure to an allergen. This distinctive reaction is often used in testing for allergies to determine which allergens trigger a reaction in a patient. When a wheal and flare reaction occurs outside the context of a doctor's office, it can be a sign that someone is about to experience a severe allergic reaction to something he or she has come into contact with, and it can be a good idea to be prepared for further symptoms such as difficulty breathing
2 pictures of wheel and flare reaction
lecture 20: 6a and 6b
What is Stat6 activated in response to ?
IL-4......during a TH2 response
scheme with level of cytokine production by T cells grown in presence of IL-4
lecture 20: 6c
talk about T-bet mouse
lecture 20: 7a
talk about drugs in treating type 1
lecture 20: 7b
what type of hypersensitivity reaction would occur against blood cells?
type 2
Draw the ABO blood groups and chart with genotype, blood-group phenotype, and antigens on erythrocytes and serum antibodies
389
what kind of Ab do we make to ABO blood group antigens?
IgM
what kind of antibodies do we make to Rh factor?
IgG
development of erythroblastosis fetalis.....2 schemes
390
type 3 hypersensitivity reaction
called arthus reaction

lecture 20: 8c

pg 392
talk about serum sickness
-immune complexes can occur which are hard for phagocytes to clear

--can cause tissue damagin type 3 reactions at various sites

-complexes cause kidney probems, arthritis and vasculitis

see page 392

lecture 20: 9b
dth stands for what?
delayed type hypersensitivity
the dth response
-type 4

394
experiment showing the importance of cytokines in DTH response
396
draw contact hypersensitivity response....what type is this?
type 4

lecture 20: 10c

397