Shared Flashcard Set

Details

III midterm
n/a
308
Health Care
Graduate
09/12/2009

Additional Health Care Flashcards

 


 

Cards

Term
what is the triple response of lewis?
Definition
3 things that classically happen in allergic reaction: vasodilation, increased vascular permeability, and binding to afferent nerve endings resulting in reactive flare
Term
what are 4 things the mast cell releases upon degranulation in hypersensitivity?
Definition
histamine (triple response of lewis), platelet activating factor (pulls in neutrophils and eosinophils), TNF-alpha (proinflammatory cytokine, increases adhesion molecules and stimulates monocytes), IL3 IL5 and CSF (maintenance cytokines, go to bone marrow and stimulate release of more eosinophils)
Term
if there is enough of a strong signal (in hypersensitivity reaction) what happens via phospholipase A2?
Definition
phospholipase A2 cleaves arachadonic acid (inner cell membrane) and either goes thru the cyclooxygenase pathway (forms prostaglandins, which are proinflammatory) or the lipoxygenase pathway (form leukotrines C4, D4 and E4, which are slow reactive substances of anaphylaxis. 1000x more potent than histamine causing bronchoconstriction, increased vascular permeability and vasodilation)
Term
give 3 examples of hypersensitivity type II reaction?
Definition
heparin-induced thrombocytopenia, RH+/- reaction with mother/fetus, induced hemolytic anemia (pcn)
Term
describe hypersensitivity type III reactions?
Definition
soluble protein ag binds with their IgG Ab --> in mid to late response the IC circulate to various places in the body (joints, kidney, skin, brain) --> complement activated and C5a is formed (neutrophil chemoattractant) --> neutrophil mediated tissue and organ damage can result (neutrophils eat and release granules --> localized damage)
Term
what are 2 examples of type III hypersensitivity reactions?
Definition
systemic lupus erythematosus and serum sickness from prolonged use of high-dose pcn
Term
what is an arthus reaction?
Definition
Ag is re-introduced into the skin (tb test, etc) --> IgG specific for that Ag binds to it and in situ IC formed with resultant inflammatory reponses (d/t complement activation and C5a pulling neutrophils into the area causing localized inflammation)
Term
Describe the Th1 mediated hypersensitivity reaction aka macrophage-meditated (subclass of type IV DTH)?
Definition
ag presented to sensitized CD4 T cells that produce Th1 cytokines (INFgamma and TNFalpha) --> produce macrophages --> swelling (ex: TB skin test) OR ag --> development of cell granulomas form in an attempt to wall off the unknown ag --> irreversible tissue damage and organ dysfunction (ex: sarcoidosis - these pts often have clinical anergy bc the CD4 cells are preoccupied with the site of active disease and cannot mount an effective cell mediated memory response to the skin test)
Term
what is clinical anergy?
Definition
when we don't respond properly to recall antigens (candida and trichophyton, both skin fungi) to which the population at large is exposed. results from immunosuppression either as a result of drugs being given or immunodeficiency
Term
what 3 types of substances are produced by Th1 cells in Th1 mediated hypersensitivity?
Definition
chemokines (macrophage recruitment to ag site); cytokines (IFNgamma activates macrophages and increases release of inflammatory mediators, IL3 and GM-CSF monocyte production by bone marrow stem cells); cytotoxins (TNFalpha and beta --> local tissue destruction, increased expression of adhesion molecules on local blood vessels)
Term
describe Th2 mediated hypersensitivity aka mast-cell mediated hypersensitivity?
Definition
mast cell signals --> late-phase response involving influx of eosinophils and Th2 lymphocytes (i.e. airway changes in chronic allergic asthma and allergic rhinitis)
Term
describe the CD8-mediated type IV hypersensitivity reaction.
Definition
when ag is cell-associated --> immunologic memory response results in CD8 destruction of cells to which ag is associated (i.e. contact dermatitis with poison ivy. exposed once --> pentadecacatechol associates with and modifies proteins in our skin cells --> modified proteins processed by dendritic cells, presented with MHC I to T-cells in local lymph node --> re-exposure to poison ivy --> memory response --> cytotoxicity to any skin cell exposed --> typical papular-vesicular skin lesion pruritic!
Term
what are 4 things that make cell injury irreversible?
Definition
any profound disturbance of the membrane function, release of lysosomes, nuclear injury that causes pyknosis, karyorrhexis, or karyolysis; lastly, increased swelling of mitochondrial membrane with vacuolization of mitochondria, cristae, and inner mitochondrial membrane and increased calcification
Term
describe coagulation necrosis
Definition
cell swelling or rupture. loss ofnucleus but preservation of cell "ghost" most commonlyhappens after hypoxic, ischemic, or chemical cell injury. grey firm mass like cooked meat.
Term
describe liquefactive necrosis
Definition
ischemia --> proteolytic digestion of lipid --> loss of original architecture. results in cystic cavities. characteristic of brain lesions and abscesses
Term
describe fat necrosis
Definition
occurs anywhere fatty (i.e. breast, pancreas, fatty tissue). enzymatic lipolytic activity --> cleaves triglycerides into glycerol and free fatty acids --> forms calcium soaps, chalky white deposits in areas of fat necrosis
Term
describe caseous necrosis
Definition
seen in well circumscribed granulomas (become filled with cheesy, amorphous, protinaceous, caseous material). typically found in the center of TB lesions. no original architecture seen
Term
describe fibrinoid necrosis
Definition
accumulation of plasma proteins --> alteration of injured blood vessels
Term
what type of necrosis is gangrene and what are the 3 different types?
Definition
coagulative necrosis, usually affects a leg that has lost its blood supply. (1)dry - ischemic necrosis of a limb, dry, dark brown to black (2)wet - saprophytic bacteria superimposed on dry gangrene causes putrefactive changes (3)gas - wet gangrene in which saprophytic bacteria are gas formers
Term
what is the function of BCL-2?
Definition
it is anti-apoptotic. prevents the release of cytochrome C from the mitochondria and prevents capase (apoptotic cascade enzyme) activation
Term
what three things induce apoptosis?
Definition
growth factor deprivation, DNA damage, and cytotoxic T cells
Term
what is anoikis?
Definition
apoptosis induced by loss of cell adhesion on the extracellular matrix (ECM provides physical scaffold on which cells adhere and the orientation context for the cells in a multicellular organism)
Term
what do p53 genes do?
Definition
they trigger a cell to undergo apoptosis (whereas BCL-2 and NF-kB favor cell survival)
Term
what are the enzymes used in necrosis? apoptosis?
Definition
necrosis - lysosomal hydrolases; apoptosis - capases
Term
what do proteosomes do in response to cell injury? if they are not successful, what happens?
Definition
unfold and refold denatured polypeptides to restore their function and reverse partial denaturation. if they cannot restore function, ubiquitin binds to the denatured protein for disposal
Term
describe dystrophic calcification
Definition
abnormal deposition of calcium salts in injured or dead tissues. the serum calcium is NOT increased. extracellular deposition with lipid accumulation. happens in vasculature and heart valves --> compromises function by narrowing arteries and limiting blood flow, valves become stiffened and may limit movement
Term
describe metastatic calcification
Definition
deposition of calcium salts in undamaged (normal) tissues when serum calcium abnormally increased. (could be d/t: hyperparathyroidism, sarcoidosis, cancer with bony metastases, hypervitaminosis D, immobilization, milk-alkali syndrome)
Term
what is the pathophysiology of fatty change?
Definition
excessive mobilization and delivery of free fatty acids to cells. inability to convert fatty acids to phospholipids. increased esterification of fatty acids to triglycerides. impaired synthesisofproteins needed for transport of secreted lipoproteins. (occurs with ischemia, hypoxia, and alcohol abuse)
Term
glycogen accumulation in cells is most often a result of?
Definition
diabetes mellitus
Term
what are the 2 main areas of excess protein production, and what does intracellular protein accumulation cause?
Definition
antibody-forming plasma cells (B-cells) and renal tubules when protein is excreted. causes compression and disruption of organelles
Term
what is anthracosis?
Definition
accumulation of carbon (coal dust) in alveolar macrophages of the lungs (d/t burning of fossil fuels) harmless condition
Term
what is silicosis?
Definition
accumulation of silica, asbestosis, silver, lead, or iron. can lead to mesothelioma (cancer)
Term
what is the effect of too much tetracycline?
Definition
chronic maternal use stains teeth and soft fetal tissue
Term
what is lipofuscin?
Definition
intracellular accumulation of lipids and protein d/t wear and tear. yellow-brown. not digestible. increases with age. seen in heart, nerve, liver. not injurious to cell structure or function
Term
what is melanin accumulation?
Definition
intracellular accumulation of melanin --> brown-black, formed i n melanocytes by oxidation of tyrosine. normally in skin, retina, leptomeninges, substantia nigra. melanomas usually produce melanin. albinos have a hereditary lack of tyrosinase.
Term
describe intracellular increase in hemosiderin and its local and systemic causes.
Definition
accumulation in macrophages usually from hemoglobin, contains iron. golden-yellow. ferritin aggregates forming apoferritin-iron complex. local cause is bruising/hemmorrhage. systemic cause is iron-overload disorders (hemochromatosis, hemolysis, excessive blood transfusions) deposition in liver, pancreas, spleen, lymph node, bone marrow. can cause fibrosis if severe.
Term
describe intracellular accumulation of hemozoin
Definition
golden-brown, comes from hemoglobin accumulating in kupffer cells in malaria
Term
describe intracelllular accumulation of bilirubin
Definition
yellow-orange to green-brown. from hemoglobin, NO iron, normal bile pigment. increase --> jaundice. retention is either d/t overproduction or inability to excrete it. product of abnormal synthesis or metabolism
Term
what are 4 examples of how free radicals can cause cell injury?
Definition
(1)metabolism of drugs, i.e. acetaminophen overdose. produces free radicals that cannot be cleared quickly enough by glutathione --> liver necrosis (2)chemicals, ie CCl4, when transformed into CCl3, becomes free radical that impairs hepatic protein synthesis --> inability to transport lipids out of cell --> rapid buildup in hepatic triglycerides --> diffuse fatty change --> necrosis in centrilobular liver where CCl4 is stored (3) lipid peroxidation, double bonds of lipids in membranes attacked by free radicals --> produce peroxides (unstable) --> autocatalytic chain reaction to membranes, organelles, and cells (4)oxidation of amino acids and ssDNA breaks
Term
describe how reperfusion injury happens and how we treat it
Definition
ischemia --> no oxygen --> no reactive oxygen species being created. during ischemia, xanthine dehydrogenase converted to xanthine oxidase, which accumulates in the cell. when oxygen is restored --> xanthine is oxidized producing ROS (does not affect cells that are irreversibly damaged, but in viable cells that have damaged mitochondria and a compromised antioxidant system are vulnerable to damage) tx: give supplemental free radical scavengers to help neutralize the ROS created after reperfusion, or give allopurinal, which inhibits xanthine oxidase altogether
Term
what IS amyloid?
Definition
abnormally folded proteins that get deposited as FIBRILS extracellularly (particularly in blood vessels and basement membranes)amorphous, eosinophilic, hyaline. they form as a result of a failure to break down abnormally folded proteins.
Term
amyloid has morphological similarity to which other 2 substances? how do we determine its amyloid?
Definition
collagen and fibrin. histologically distinctive under light microscopy with affinity for special dyes ie. congo red, turns green under polarized light. also, they have beaded nonbranching fibrils and beta-pleated sheet under xray crystallography
Term
describe immunocyte dyscrasia with amyloidosis (primary amyloidosis)
Definition
most common form of amyloidosis. occurs in patients who synthesize abnormal amounts of a specific immunoglobulin (monoclonal gammopathy), such as multiple myeloma (occurs 5-15%) and other monoclonal B cell plasma dyscrasias. **the fibroid that gets deposited is amyloid light chain (AL) with lambda>kappa
Term
describe reactive systemic amyloidosis (secondary amyloidosis)
Definition
**protein causing fibril: AA (amyloid associated protein)non-immunoglobulin, synthesized in liver** (precursors of AA are SAA and alpha-1 globulin)occur in cases of drawn-out cell injury ie chronic injury and chronic inflammation. frequent in rheumatoid arthritis, inflammatory bowel disease, heroin use, renal cell carcinoma, hodgkins, tb, osteomyelitis
Term
describe hemodialysis-associated amyloidosis
Definition
**protein fibril: B2-microglobulin** deposits in synovium, joints, and tendon sheaths of long-term dialysis patients
Term
what fibril involved in hederofamilial amyloidosis?
Definition
AA or transthyretin. recurring episodes of inflammation, rare, characteristically in black males in conjunction with cardiac failure
Term
what fibril associated with localized amyloidosis?
Definition
AL. localized form of immunocyte-derived amyloid
Term
fibril involved in amyloid of aging?
Definition
transthyretin
Term
fibril associated with alzheimers disease?
Definition
beta-amyloid (in blood vessels in the brain)
Term
describe the causes of nutritional edema
Definition
(due to decrease in intravascular osmotic pressure as a result of a profound decrease in albumin) (1)liver failure --> decreased albumin synthesis by liver (2)nephrotic syndrome (glomerular capillary damage) --> loss of albumin through kidney (3)starvation --> profound decrease in protein intake
Term
describe the differences b/w transudate and exudate
Definition
transudate= passive, hydrostatic, sterile, clear, low protein content, few cells, usually indicates heart failure. exudate=active, secondary to inflammation, infection, tumor etc. not sterile (has microorganisms), cloudy, high protein content, numerous cells, usually indicative of infection or tumor
Term
briefly describe the process of hemostasis
Definition
(clotting after vessel is severed) vessel severed --> transient, insignificant arteriole constriction --> interruption of laminar blood flow --> platelets leave vessel and adhere to extracellular matrix/collagen via binding to von willebrand factor --> platelet release reaction (platelets secrete ADP, thromboxane A2, which mediate platelet to platelet adhesion and aggregation) --> form primary hemostatic plug (reversible) --> platelets fuse --> activation of coagulation cascade --> generation of thrombin --> fibrinogen --> fibrin, which stabilizes the clot and makes the secondary hemostatic plug --> endothelium regenerates, clot undergoes fibrinolysis via plasmin,e tc
Term
what is virchows triad?
Definition
3 major risk factors that can predispose someone to thrombus formation: (1)endothelial injury (2)disturbances of blood flow (stasis in veins, turbulence in arteries) (3)disturbances in the composition of blood (hypercoagulable states, increased viscosity of the blood, sickle cell)
Term
how is the morphology of arterial thrombi different from venus thrombi?
Definition
arterial thrombi resembles a platelet-fibrin plug (usually d/t endothelial injury, if it occurs in the left heart, will have systemic effect); venous thrombi develops laminations (lines of ZAHN, composed of fibrin and platelet deposits alternating with layers of erythrocytes) d/t stasis, will end up in lung usually
Term
what are the 3 most common sites of pulmonary thromboemboli?
Definition
deep veins of the leg, pelvic plexuses (periprostatic, utero-ovarian), brachial veins or right atrium (rare)
Term
what part of the immune system responds to typical acute extracellular bacterial infection?
Definition
humoral immunity (antibody-mediated) ie. opsonization, complement fixation, bacterial immobilization, adhesion blockage, toxin neutralization
Term
phagocytes have 2 important receptors on their surface. what are they?
Definition
Fc receptor (for opsonization of encapsualted bacteria especially) and C3b receptor (for complement fixation)
Term
people with defects in the lytic pathway of complement have markedly increased susceptibility to what bacterial species?
Definition
neisseria (ie repeated instances of meningitis or sepsis caused by N. meningitidis)
Term
what 2 processes are considered bacterial neutralization?
Definition
anti-flagellar antibodies (flagella can be antigen) immobilize bacteria; antibodies against bacterial surface adhesins can block their adhesion to host tissue surfaces
Term
how does toxin neutralization work against bacterial infection and why is it important
Definition
specific antibody for a toxin can neutralize it by sterically blocking their active sites or by allosteric interaction - is an immediate effect. important bc killing toxigenic bacteria has no effect on the toxin they already made/released.
Term
what defenses are best used against an intracellular bacterial infection?
Definition
CD8 cytotoxic T lymphocytes and macrophage activation by CD4 helper T cells (TH1)
Term
how does the body defend itself against infection caused by opportunistic environmental molds?
Definition
IR is largely irrelevant. they primarily infect patients who have defective neutrophil numbers or activity (leukemia, diabetes).
Term
what role does cell-mediated immunity have against molds?
Definition
CMI necessary to control infections and may itself contribute to most of the sx. in its absence the sx are different and disease takes more progressive course. these molds are similar to mycobacteria
Term
whats our defense against helminths?
Definition
TH2 response - release of toxic granule contents from eosinophils and mast cells, targeted at helminths by immune IgE molecules. may eliminate worms or decrease the ultimate worm burden as infection is established. (more effective in smaller larvae. no real defense against established helminths)
Term
major cytokines of inflammatory rxn?
Definition
IL1, TNF-alpha, IL6
Term
describe INF (alpha and beta)
Definition
products of any cell infected by a virus; transmissable from cell to cell; shuts down cell translation (viral and host proteins not being made); nonspecific; pro-TH1
Term
What do NK cells do?
Definition
they kill virally infected cells. they have activating and inhibiting receptors on cells. healthy cells make inhibitors (ie MHC I, virally infected cells lose their MHC I)
Term
explain original antigenic sin
Definition
for some pathogens (ie influenza) epitopes are changing all the time (phase variation) so our IR to it via memory cells gets weaker and weaker. suppression of IgM in secondary response bc circulating IgGs from 1st exposure coat the ag and prevent a robust response by naive B cells (therefore we dont create NEW memory cells for the NEW epitopes that are mixed in). the ag does trigger a robust memory B cell response to the 'old' epitopes. naive B cells shut down bc new epitopes are physically linked to the 'old' epitopes that have already been recognized by the b cell.
Term
describe the herd immunity formula for how many people must be vaccinated?
Definition
(1-1/R)/(vaccine efficiency) ie we need to decrease the # of susceptible individuals to less than the reciprocal of R.
Term
briefly describe the lac operon
Definition
absence of lactose --> lac repressor diffuses thru cytosol and binds to lac operator, prevents initiation of RNA polymerase at lac promoter. lactose present --> conformational change lactose to allolactose --> allolactose binds to repressor, changes conformation of the repressor --> repressor falls off of the DNA --> RNA polymerase free to move along lac operon and mRNA can be transcribed to yield genes that allow lactose to be digested
Term
briefly describe catabolite repression
Definition
glucose high --> cAMP low. glucose low --> cAMP high --> cAMP binds to CAP protein (catabolite activator protein) --> CAP protein binds to CAP binding site near promoter --> activates transcription perhaps by increasing the affinity of the promoter for RNA polymerase. (lac operon requires both de-repression via lactose and activation via CAP for transcription to occur)
Term
what does the DtxR regulate? and how is the DtxR toxin and operon system an example of end-product inhibition?
Definition
DtxR regulates all operons dealing with Fe levels (>18 different operator sequences controlled by this toxin) controls synthesis of iron binding proteins (siderophores), iron transport, etc. it is an ex of end product inhibition bc the presence of iron shuts off transcription frmo operons and genes required for iron acquisition and utilization
Term
how does the presence of iron activate the operon repressor?
Definition
if iron is present --> will bind to the repressor and keep the repressor in its proper conformation necessary for it to keep repressing. iron absent? --> iron falls off receptor --> repressor assumes an improper/irregular shape --> repressor falls off the DNA
Term
describe the two-component regulation system via the example of bordetella pertussis
Definition
two proteins involved - BvgS (sensor protein, detects environmental changes in temp) and BvgA (DNA binding protein, functions as activator of the transcription of virulence genes). at 37 degrees --> BvgS autophosphorylates --> activates BvgA --> activates transcription of virulence genes
Term
what is an insertion sequence and what does it do?
Definition
small sequence containing a gene coding for a single enzyme (recombinase or transposase), which cuts DNA and allows it to come off and be moved somewhere else. bounded by inverted repeats. may result in extreme polar effects, constitutive gene expression, or inactivation and/or rearrangement of nearby genes (contributes to genetic plasticity)
Term
describe phase variation controlled by DNA inversion (salmonella flagella!)
Definition
flagellins H1 and H2 mutually exclusive. phase 2: H1 gene silenced by repressor protein (rh1) formed along with H2 flagellin. phase 1: inversion of DNA segment catalyzed by a recombinase (hin) encoded by it --> loss of promoter for H2 and the repressor for H1 --> H1 now expressed. (further inversions switch system back and forth between those two phases)
Term
describe class I (composite) transposons
Definition
genes flanked by 2 IS elements (genes can encode for drug resistance, etc.) ends of transposon can transpose separately. can be nonreplicative or replicative
Term
describe class II (complex) transposons
Definition
contain 1 or more genes within a single IS element. must transpose as a unit. (necessarily replicative)
Term
how does phase variation work?
Definition
an IS element in bacterial genome can flip its orientation by the recombinase (random) and can turn on or turn off a series of genes. (if protein subject is on the surface of the cell --> subject to antigenic selection ie. influenza with new epitopes all the time)
Term
for bacterial conjugation, what direction is the dna transferred to the recipient cell? and which secretion system is employed?
Definition
5-->3 direction. type IV secretion system
Term
describe generalized transduction
Definition
occurs during the lytic phase of bacteriophage transduction. observed with bacterial viruses that dont completely degrade host DNA --> packaging of host DNA into viral capsids
Term
what's abortive transduction and what's the consequence/result?
Definition
when host DNA that was not efficiently broken down gets packaged into viral capsids --> infects another bacterial cell --> host DNA gets transferred but not integrated into new cell's DNA --> persists unreplicated for a few generations --> genetic info gets expressed in recipient cell --> formation of minute colonies under selective conditions
Term
describe specialized (restricted) transduction
Definition
occurs during the lysogenic phase of bacteriophage infection. (when conditions favor lysogenic state, phage DNA integrates into specific site in bac chromosome --> environmental conditions cause phage to excise its dna from host and enter lytic cycle) --> if excision not precise, bacterial DNA can be excised with the phage DNA --> if packaged into a capsid, that transducing particle may or may not be defective depending upon whether essential or non-essential phage genes were left behind
Term
type III secretion systems are widespread among gram____ bacteria? what processes do they facilitate?
Definition
gram NEG. allow bacteria to inject virulence proteins into cytosol of eukaryotic host cells. allow bacteria to invade non-phagocytic cells, inhibit phagocytosis, down/up regulate inflammatory response, induce apoptosis, modulate intracellular trafficking
Term
how do bacteria force non-phagocyte cells to phagocytose them?
Definition
gram NEG bacteria secrete invasin into eukaryotic host cell --> invasin makes change in cell surface (introduced by type III secretion system) --> invasins send in other materials to rearrange cellular actin --> cause changes to host cell cytoskeleton --> engulfment by host cell
Term
how does bacteria inhibit phagocytosis?
Definition
bacteria adheres to phagocyte by bundle forming pili --> secretes virulence factors into host cell (ie Tir) --> signal transduction --> actin recruitment --> rearranges cell surface into pedestals and actin brings Tir up to surface of pedstals so e coli can be more tightly bound --> pedestal formation inhibits phagocytosis
Term
explain the mechanics of flagella movement
Definition
related to type III secretion systems. drives its force by proton flow through proteins in the envelope --> counter clockwise rotation --> energy runs low --> motion stops and reverses (cell tumbles)--> new surge of energy? --> reversal to counterclockwise
Term
what are 3 ways bacteria can get iron from the host celL?
Definition
1. release siderophores which bind to receptors on bacterial surface --> iron taken up by cell from lactoferrin/transferrin/ferritin/hemin 2. bacteria binds directly to cellular iron chelator proteins 3. bacteria excretes proteases that kill host cells --> iron released and available for uptake
Term
what are 2 ways LPS can be modified by pathogens to affect its interaction with complement?
Definition
1. attachment of sialic acid residues to the LPS O antigen --> prevents formation of C3 convertase 2. LPS with long, intact O antigen side chains can prevent effective MAC killing
Term
how do bacteria "neutralize" antibodies?
Definition
some bacteria release soluble antigenic surface components (ie protein A) into surrounding fluids by gram NEG bacteria --> protein A in soluble form binds to Fc region of IgG --> agglutinate --> partially inactivates IgG. surface-bound protein A --> binds IgG in wrong orientation to exert its antibacterial activity (Fc region, not Fab)
Term
describe type I secretion system
Definition
3 proteins form transmembrane channel, driven by ATP-binding cassette. proteins lack a signal peptide.
Term
describe type II secretion system
Definition
(2-step secretion) uses "general secretion pathway" to move proteins into periplasm --> protease cleaves N-terminus signal peptide upon transfer --> N-terminal signal sequence seen in periplasm --> step 2: 14 accessory proteins move secreted protein across the outer membrane. require ATP.
Term
describe type III secretion system
Definition
(for contact-dependent secretion) involves 20 proteins, including an ATP-binding protein and chaperone protein specific for each different protein to be secreted. --> direct injection of secreted protein into cytoplasm of mammalian host cell
Term
describe type IV secretion system
Definition
used to transfer DNA from donor cell to recipient cell during conjugation. plays crucial role in spread of antibiotic resistance genes
Term
type V secretion
Definition
auto-transporter - no helper protein required for translocation through outer membrane (secreted protein itself serves this function). is a 2-step system which requires the general secretion system
Term
endotoxin vs exotoxin - which is denatured by boiling?
Definition
exotoxin
Term
endotoxin vs exotoxin - which can be treated to form a toxoid?
Definition
exotoxin
Term
endotoxin vs exotoxin - which has a higher degree of specificity?
Definition
exotoxin
Term
endotoxin vs exotoxin - which one is more pyrogenic?
Definition
endotoxin
Term
where are the genes for exotoxins usually found?
Definition
plasmids or temperate phages (can be mobilized to nontoxigenic bacteria)
Term
give a few examples of exotoxins that work outside the cell (not intrinsically toxic).
Definition
hyaluronidase, collagenase, DNAase, streptokinase, fibrinolysin, coagulase
Term
give 2 examples of exotoxins that work on the cell surface.
Definition
lipases, pore-forming toxins
Term
describe the mechanism of lipase exotoxins (via example of lecithinase or alpha toxin)
Definition
intended to lyse phagocytic cells and increase the availability of nutrients to bacteria. cleaves the hydrophilic head of lipid membrane components --> destabilizes the membrane --> membrane rupture. at high concentrations, alpha toxin --> massive degradation of phosphatidylcholine --> membrane disruption. small amounts of toxin --> limited hydrolysis of phosphatidylcholine (generate diacylglycerol) --> signals transduction pathways leading to uncontrolled production of cellular mediators (adhesion molecues, IL8, TNFa, platelet-activating factor..all of which contribute to increased vascular permeability and edema)
Term
describe mechanism of pore-forming toxin action
Definition
toxins bind to cell surface --> oligomerize to form a pore (homogeneous vs heterogenous) --> pore partially inserts into cytoplasmic membrane --> depletion of ions and metabolites --> allow H20 to enter cell --> cell bursts
Term
describe the mechanism of toxins that act inside the cell
Definition
A-B toxins. (can be on same polypeptide or on 2 different ones bridged by disulfide bonds). B moiety gets the toxin bound to and taken up by the cell --> A moiety activated by proteolytic cleavage or reduction of disulfide bridges --> act as ADP phosphoribosyltransferases, transfer ADP from NAD to a particular target protein in cell --> kill target cell or elevate/depress normal cell function w/o killing the target cell --> can move onto another host cell
Term
describe how the diptheria toxin gets into a cell and what it does inside the cell
Definition
(max levels of toxin when iron levels low) B component binds to specific receptors and cleaved into A and B portions, which remain associated via disulfide linkages --> endocytosis --> A component released by reduction of disulfide linkage by the acidification of the cell interior --> toxin acts upon EF-2 via ADP ribosylation of a modified histine residue (EF-2 is a GTP hydrolase required for ribosomal translocation)--> EF-2 modification inhibits protein synthesis --> local tissue necrosis
Term
describe the pathophysiology of diptheria exotoxin action (ie what happens after cell protein synthesis is inhibited and tissue becomes necrotic)
Definition
affects superficial layers of skin lesions or usually respiratory mucosa. dense necrotic coagulum forms gray-brown adherent pseudomembrane (removal difficult and reveals bleeding submucosa)-->paralysis of palate and hypopharynx. toxin absorption can lead to kidney tubule necrosis, thrombocytopenia, cardiomyopathy and demyelination of nerves.
Term
describe the cellular mechanism of cholera toxin
Definition
drink contaminated water --> vibrio comes into contact with gut --> B unit binds with high affinity to GMI ganglioside (contact type III secretion) --> toxin-GMI complex forms --> conformational alteration of holotixn allows presentation of A subunit to cell surface --> A component taken up via endocytic vesicles--> A component finds G protein on cAMP kinase system and ribosylates it(inactivates it, locking adenylate cyclase in "on" mode) --> uncontrolled release of cAMP --> epithelial cells move large amt of fluid across intestinal membrane into gut lumen
Term
describe the cellular mechanism of tetanus toxin action
Definition
wound contamination by bacterial spores --> spread through lymphatics and vascular system --> enter myoneuronal junctions by binding to ganglioside receptors and getting internalized --> migrates thru CNS by retrograde axonal transport --> B chains bind specific receptors on neuron membranes and A chain is the zinc endopepsidase --> cleaves synaptobrevin (on the vesicle, forms assoc w/SNAP & syntaxin on membrane surface and cause vesicle to come down to membrane and fuse and release) --> no assoc of vesicle with membrane --> no release of inhibitory neurotransmitters (GABA and Gly) --> generalized contraction/rigid paralysis
Term
describe the cellular mechanism of botulinum toxin
Definition
disease results from ingestion of the toxin (true intoxication) --> once across the gut toxin carried in the blood to neuromuscular junctions --> binds to ganglioside receptors and is taken up --> toxin causes presynaptic block by cleaving synaptobrevin and/or syntaxin or snap --> blocks release of acetylcholine --> irreversible flaccid paralysis (death thru respiratory arrest)
Term
what is the basic structure of the anthrax toxin?
Definition
AB toxin. single B subunit (protective antigen) and 2 alternative A subunits (edema factor and lethal factor)
Term
4 structural domains of anthrax toxin B subunit (PA or protective antigen)
Definition
domain 1 - proteolytic activation site; domain 2 - B-barrel core containing large flexible loop for pore formation; domain 3 - involved in heptamer formation; domain 4 - binds to anthrax toxin receptor
Term
4 domains of anthrax toxin's lethal factor LF (one of the A subunits)
Definition
domain 1 - PA binding domain; domain 2 - forms binding pocket for substrate peptide; domain 3 - restricts access to binding pocket; domain 4 - catalytic center
Term
what does EF (edema factor) of the anthrax toxin do?
Definition
calcium-calmodulin dependent adenylate cyclcase that causes dramatic increases in intracellular concentrations of cAMP
Term
what does LF (lethal factor) of the anthrax toxin do?
Definition
zinc-dependent protease, cleaves mitogen-activated protein kinase kinases in amino-terminal regions. early in infection suppresses immune cell and cytokine responses --> promoting bacterial outgrowth. later in disease induces cytokine independent shock-like death associated with anthrax.
Term
how is anthrax toxin introduced into the cell?
Definition
PA (B component) binds to cell surface anthrax toxin receptor --> PA cleaved by furin or furin-like protease into PA20 (amino terminal) and PA63 (receptor-bound carboxy terminal) --> PA63 internalized and directed to endosomes --> low pH in endosome induces conformational change in PA63 heptamer --> formation of cation-selective channel --> translocation of EF and/or LF across endosomal membrane
Term
describe the cellular mechanism of the shigella toxin
Definition
A subunit and 5 molecules of B subunit. B subunits bind to G3b (component of cell membrane) --> enters cell --> A subunit modifies 28s ribosomal RNA and inactivates protein synthesis --> cell death (toxin acts on vascular endothelium --> breakdown of lining --> hemorrhage) first response = bloody diarrhea. effective in GI tract, kidney and lungs
Term
how does LPS alert the body to the presence of gram-negative organisms?
Definition
sets off series of alarm reactions. fever (via release of IL-1 and TNF by mononuclear phagocytes), activation of complement (via alternate pathway, and anaphylatoxins result in vasodilation and inflammation), activation of macrophages, stimulation of B-lymphocytes (at high concentrations can induce shock and disseminated vascular coagulation)
Term
what are the 2 stages of pertussus infection?
Definition
catarrhal stage (nonspecific cold-like symptos, very contagious, organism present in high concentrations in URT secretions, easily identifiable via culture) and paroxysmal stage (more severe and frequent bouts of coughing, organism concentration dramatically decreased, coughing interferes w/normal breathing can cause hypoxia, seizures, coma, vomiting, weight loss)
Term
how do we test for bordetella pertussis?
Definition
need to plate on growth medium specifically formulated for b. pertussis (fastidious) during catarrhal stage. no test or culture is sensitive during paroxysmal stage therefore must suspect it clinically and order specific tests early on. serology useful in hindsight.
Term
what treatment do we administer for pertussis?
Definition
tx helps shorten period of transmission. erythromycin, macrolides, doxycycline, sulfonamides. problem with tx is that damage to respiratory epithelium is irreversible until epithelium regenerates so clearing of the infection doesnt do much about the damage or the sx.
Term
how does b. pertussis cause a cough?
Definition
b. pertussis has tropism for ciliated respiratory epithelium --> binds to and disables ciliary motion --> failure to properly clear respiratory secretions and also kills epithelial cells
Term
how do the bacterial toxins pertussigen and invasive adenylate cyclase work in pertussis?
Definition
pertussigen is AB toxin. on cell surface of bacterial cells serves as adhesion to tracheal epithelium. can induce lymphocytosis and interferes w/normal functions of phagocytes, lymphocytes, and tracheal epithelial cells --> epithelial damage, mucous accumulation, blunting of immune response. toxin is an ADP ribosylase, ribosylates inhibitory G-protein which normally deactivates adenylate cyclase --> cAMP accumulates --> different effects in different cell types
Term
what does tracheal cytotoxin do in pertussis?
Definition
decreases ciliary function and in high enough doses induces epithelial cell production of proinflammatory mediators (IL-1 and NO) that contribute to killing epithelial cells
Term
what does dermonecrotic toxin do in pertussis?
Definition
vasoconstrictive effects. inactivates rho-GTPase, resulting in constitutive signaling by rho GTP-binding protein. helps kill epithelial cells
Term
what does filamentous hemagglutinin do in pertussis?
Definition
lipo-oligosac. func as adhesins together with surface-bound pertussis toxin. bacteria binds to cells with both adhesins --> adhesins x-link cilia --> mechanically disable cilia and disable effective mucous clearance
Term
what is the main branch of immune sys responsible for recovery from infection of pertussis?
Definition
humoral immunity. ab titer rises with decline of recoverable bacteria on mucosal surface.
Term
what are the 3 mycoplasmal species that are unambiguously associated with human disease?
Definition
mycoplasma pneumoniae (primary atypical pneumonia); mycoplasma hominis (PID, pyelonephritis); ureaplasma urealyticum (NGU)
Term
compare primary atypical (mycoplasma) pneumonia to classical lobar (strep) pneumonia
Definition
classical - looks like alveolar infection with whole lobe consolidation (on xrays), respiratory complaint = chief compaint. atypical - inflammatory infiltrate of parenchyma has patchy appearance on xrays, much less of a severe infection, more likely see general systemic illness complaints
Term
what causes the cough in primary atypical pneumonia?
Definition
inhibition of ciliary function--> poor mucous clearance --> cough. no specific inhibitory toxins have been identified, but evidence for ciliary toxicity of bacterial metabolic products such as hydrogen peroxide. little structural damage to epithelium.
Term
how helpful is tx with antibiotics for atypical pneumonia?
Definition
without tx, infection is self-limited, resolves gradually over several weeks. with tx (macrolides, tetracyclines) can shorten sx, but does not result in faster clearance of organism from body bc they multiply intracellularly.
Term
what are cold agglutins and how do we test for them?
Definition
common autoimmune mechanism symptomatic of atypical pneumonia. formulation of auto-ab against patient's RBC. draw blood and anticoagulate and refridgerate --> agglutination (reversible, if we warm blood back up, will go back into suspension) can cause hemolytic anemia
Term
give some examples of how sx in mycoplasma pneumonia moreso come from host immune system than bac toxins.
Definition
no toxins identified. rash and polyurithritis (d/t circulating IC and type 3 hypersensitivity rxn); auto-immune like phenomena aka cold agglutins, hemolytic anemia (bacteria mimic host proteins aka they share epitopes, so we accidentally mount our IR against self-proteins thinking theyre bacterial)
Term
streptococci metabolism?
Definition
aerotolerant
Term
staphylococci metabolism?
Definition
true facultative
Term
clostridia metabolism?
Definition
anaerobic spore-forming bacilli
Term
3 ways to distinguish strep species from one another?
Definition
1. lancefield groups (group carbohydrate antigens - primarily for subtyping of beta-hemolytic streptococci) 2. hemolysins (B, A) 3. antigenic specificity of M protein for subtyping of group A streptococci (virulence factor involved in resistance to opsonization)
Term
where do the endospores for clostridium form?
Definition
fecally-contaminated soil - major constituent of flora of some farm animals (only minor constituent of normal human flora)
Term
are streptococci hardy or fastidious? what do they require to grow?
Definition
fastidious. require blood agar and elevated atmospheric CO2
Term
how do we differentiate between enterococci and streptococci in lab?
Definition
enterococci are hardier organisms and can tolerate high salt. bile salt can be used to differentiate them in lab.
Term
how is pneumococci classified from other streptococci?
Definition
presence of capsular polysac antigens --> pneumococci
Term
what are some common infections of skin and throat caused by strep pyogenes?
Definition
impetigo, pyoderma, pharyngitis (strep throat)
Term
what are systemic toxinoses caused by strep pyogenes?
Definition
scarlet fever, streptococcal toxic shock-like syndrome
Term
what are some autoimmune sequelae caused by strep pyogenes?
Definition
acute rheumatic fever, post-streptococcal glomerulonephritis
Term
what are 3 ways streptococcus can infect deep tissue?
Definition
1. cellulitis of deeper tissue can result from extension of superficial skin infections (impetigo) --> erysipelas, myosistis 2. cellulitis of deeper tissue by inoculation by puncture wound 3. hematogenous spread (presumably from throat) after blunt trauma to affected area that injures muscle w/o penetrating skin (changes in circulation and/or vascular permeability favor colonization by circulating bacteria)
Term
what are the main IR players in resolution of streptococcal infections?
Definition
phagocytosis - ability to make opsonizing antibody and to activate complement (thick cell wall of gram + organisms makes strep naturally resistant to complement MAC) and field phagocytes. specific humoral immunity (esp to M protein) promotes opsonization and provides relative protection.
Term
what are the 3 most important virulence mechanisms for strep?
Definition
1. adherence to epithelia 2. elaboration of locally acting exotoxins 3. toxin-mediated and non-toxin mediated evasion of phagocytes
Term
what are strep's major adherence proteins?
Definition
protein F (key adhesin) binds to fibronectin (ubiquitous extracellular matrix protein). antibodies against protein M block mucosa adherence, but only bc these ab sterically block the protein F pilus while bound to the M protein pilus. lipoteichoic acid cooperates in adherence also.
Term
what do streptolysin O and S do to increase strep virulence?
Definition
cytolytic toxins. (streptolysin O = oxygen labile, diffusible porin; streptolysin S = oxygen stable, bound to bacterial cell surface) general cytotoxins, lyse tissue cells and immunocytes (leukocidins) these are the hemolysins that make strep B-hemolytic
Term
what does streptokinase do to promote strep virulence?
Definition
cleaves plasminogen to plasmin (plasmin is fibrinolytic) --> counteracts IR deposition of fibrin which otherwise would lead to walled-off abscesses --> makes strep a spreading infection
Term
what does hyaluronidase do to promote the bacterial virulence of strep?
Definition
classic "spreading factor" digests extracellular matrix of connective tissue and contributes to spread of infection along tissue planes
Term
how does DNAase promote bacterial virulence in strep?
Definition
pus is viscous bc released genomic DNA of lysed neutrophils and immunocytes has high molecular weight --> DNAase attacks/nicks DNA --> decreases viscosity --> bacteria can move more easily through pus (otherwise trapped for WBC attack)
Term
when is retrospective seradiagnosis useful?
Definition
when you're worried about sequelae after infection is over (i.e. in strep we worry about glomerulonephritis and rheumatic fever)
Term
why is phagocytosis the main defense against strep?
Definition
thick gram+ cell wall makes it resistant to complement MAC. phagocytosis is dependent on alternate pathway of complement or classical pathway + specific abs. strep has not evolved mechanisms for intracellular survival and is rapidly killed after phagocytosis by neutrophils or macrophages.
Term
what are strep's means of avoiding phagocytosis?
Definition
1. avoid trapping in an abscess 2. capsules (dubious importance) 3. M protein 4. c5a peptidase 5. protein G 6. superantigen exotoxins
Term
how does c5a peptidase help strep avoid phagocytes?
Definition
it inactivates phagocyte binding of c5a, which would otherwise strongly activate the phagocytes for binding and phagocytosis of c3b coated bacteria
Term
how does M protein help strep avoid phagocytosis?
Definition
M protein binds and locally concentrates factor H, soluble negative regulator of the alternate pathway C3 convertase --> inhibits deposition of major opsonin C3b on cell surface. (mimics natural binding of factor H to host cells to protect them from alternate complement lysis) destruction of bacteria by classical pathway and specific abs still works though!
Term
how does protein G help strep avoid phagocytosis?
Definition
protein G is a surface Fc receptor. thought to cloak the bacterial surface in self-antigens and sterically block immunogenic or effector functions of immune recognition
Term
what are the systemic toxins of strep pyogenes and what do they do?
Definition
streptococcal pyrogenic exotoxin A-E (erythrogenic toxin)--> induce lymphocytes to release inflammatory lymphokines (TNFa, IL1, IL6) --> fever, vasodilation via NO, vascular permeabilization, coagulation
Term
how do the streptococcal pyrogenic exotoxins (spes) induce lymphokine production?
Definition
they function as superantigens --> immediate strong general stimulation of immune system --> depletes components necessary for optimal specific IR, leads to a lot of pointless systemic inflammation --> immune paralysis. superantigens also synergize with polyclonal activators acting through other receptors like LPS from intestinal flora (LPS directly stimulate monocytes and macs to release inflammatory mediators)
Term
sx of scarlet fever?
Definition
confluent red macular rash of trunk that may spread to extremities, with fever, occuring in the course of strep throat
Term
sx of streptococcal toxic shock like syndrome?
Definition
sudden onset in the course of an invasive infection with bacteremia --> fever, profound hypovolemic shock secondary to vasodilation and capillary leakage --> multisystem failure
Term
what does the frequency, manifestation, and severity of systemic strep pyogenes toxinosis depend on?
Definition
what spes and M proteins are present together in the strains infecting a given population and the degree of immunity to those proteins that are present in the population
Term
discuss how someone comes to have acute rheumatic fever after having a resolved strep infection and what the sx are.
Definition
occurs several weeks after acute group A strep inf (usually strep throat) --> multisystemic inflammatory disorder affecting joints, skin, blood vessels, NS, heart - myocardial damage, permanent valvular defects. only appears after infections with a subset of M protein subtypes, some of which have been shown to cross-react with cardiac epitopes (type II hypersensitivity) --> x-reactive immune response to self
Term
discuss post-strep glomerulonephritis
Definition
occurs really quickly after group A strep inf (usually after skin inf) --> manifests as acute renal insufficiency --> resolves completely (may progress to chronic renal disease in adult pts). limited to certain M protein subtypes (nephritogenic strains). probably d/t immune complex disease (type III hypersensitivity)
Term
what is the significance of strep bovis (group d)?
Definition
found in blood cultures taken from febrile patients, statistically assoc with concurrent colon carcinoma (should prompt a search for it)
Term
what do enterococci most commonly cause?
Definition
autoinfectious UTI, and in compromised patients, major threat for generalized sepsis (major normal flora of bowel)
Term
subclassification of staph aureus strains is done by?
Definition
phage typing - members of diff phage groups are susceptible to inf by diff spectra of bacteriophages
Term
what are the systemic toxinoses of staph aureus?
Definition
toxic shock syndrome, scalded skin syndrome, and staph food poisoning
Term
what do alpha, beta, gamma, and delta hemolysins do to promote staph aureus virulence
Definition
lyse RBC, are cytotoxins, allow spread through tissue and kill immunocytes (alpha hemolysin is staph aureus's most important virulence factor)
Term
how does coagulase toxin contribute to staph aureus virulence
Definition
promotes deposition of fibrin to form abscess wall (denies access to IR)
Term
how does staph aureus survive IR?
Definition
can survive in and possibly kill phagocytes, surface protein A binds Fc portion of host ab molecules --> coats surface of bcteria with self-antigens --> cloaks bacteria against immune recognition; teichoic acid = soluble decomplementation antigen, results in local depletion of early complement components
Term
describe the systemic exotoxin exfoliatin
Definition
promotes exfoliation in scalded skin syndrome by weakening the cell-cell adhesions between epidermal cells (also pyrogenic, might be superantigens)
Term
describe the systemic toxin enterotoxin (for staph aureus)
Definition
dual mode of action: increase fluid secretion in small int --> diarrhea. act on vomiting center in brain via small bowel neural receptors, have superantigen activity
Term
describe pyrogenic exotoxins (systemic toxin of staph aureus)
Definition
act by superantigen mechanism, includes toxic shock syndrom toxin 1 (causes fever, vasodilation, shock in extreme cases)
Term
what is clostridia's characteristic pattern of metabolic rxn?
Definition
hemolytic, produces frothy, foul-smelling cultures in liquid media --> copious production of gases as fermentation product = virulence factor bc gas released physicall disrupts tissue
Term
what is clostridia's major locally acting toxin?
Definition
alpha-hemolysin (lecithinase, phospholipase C) --> splits phosphatidylcholine in cell membranes --> disrupts the membrane
Term
describe how we can become infected by clostridium difficile
Definition
causes pseudomembranous colitis, frequently serious and sometimes fatal complication of antibiotic therapy. (normal intestinal flora, can overgrow and cause disease when high percentage of competing flora are killed by antibiotic to which cl. difficle is resistant)
Term
briefly describe some characteristics of rickettsiae. (staining? method of reproduction? means of obtaining atp? how spread? what 3 diseases do they cause?)
Definition
do not stain positive or negative by grams stain; reproduce by binary fission; can generate ATP from their own metabolism but also have systems for import of host cytosolic ATP; are zoonotic and spread by vectors; cause epidemic typhus, rocky mountain spotted fever, ehrlichiosis; mostly responsible for chronic, indolent infections (not acute)
Term
how is the typical rickettsiae infection initiated and carried out on the cellular level? (typical as in not ehrlichosis)
Definition
inf initiated at site of vector bite --> rickettsiae invade endothelial cells by induced phagocytosis --> replicate locally for extended incubation period --> once inside cell, induce lysis of phagosome and replicate in the cytoplasmic compartment --> escape from phagosome by gross or localized cell membrane lysis --> eventual seeding of endothelium at distant sites with sx production
Term
explain why we might see an eschar at the site of local replication
Definition
intracytoplasmic multiplication --> presentation of rickettsial antigens on class I MHC --> acute necrotic response mediated by cytotoxic T lymphocytes --> formation of black necrotic scab
Term
what ticks transmit rocky mountain spotted fever?
Definition
hard ticks - wood tick in the west and the dog tick dermacentor in the east
Term
describe how rocky mountain spotted fever progresses from local infection to whole body
Definition
tick bite --> local multiplication with eschar formation --> cell to cell spread via filopodia (local cell membrane lysis) --> hematogenous spread to whole body.
Term
what are the sx of rocky mountain spoted fever?
Definition
sx referable to vascular damage: rash (distal to axial, extends to palms and soles; macular at first from vasodilation then petechial from capillary leak), fever, altered mental status, cardiac arrhythmias, .....
Term
what is mortality rate and cause in rocky mountain spotted fever? what is the going tx??
Definition
25% untx'd, from organ failure and shock. doxycycline
Term
how is epidemic typhus spread?
Definition
(all rickettsiae inf are dead-end in humans, except this one) rickettsia prowazeckii, spread human to human thru feces of the body louse which gets scratched into skin.
Term
describe the sx of epidemic typhus
Definition
no eschar, rash (macular or papular, sometimes petechial; rash axial to distal and palms and soles spared); vasculitic nodules can occlude small vessels resulting in distal gangrene of digits, nose or ears; fever; severe headache; CNS sx (lethargy, coma)
Term
what's brill's disease?
Definition
occasionally, after typhus inf, clinical latency and disease may return years to decades later
Term
what 2 species are responsible for ehrlichosis
Definition
ehrlichia chaffeensis (mainly infects monocytes and macrophages) and anaplasma phagocytophilia (mainly infects neutrophils)
Term
what are the ticks that transmit e. chaffeensis and a. phagocytophilia?
Definition
e.chaffeensis = lone star tick; a.phagocytophilia = ixodes tick
Term
what makes ehrlichosis inf different from RMSF or typhus?
Definition
infections take place in the endosomal compartment of the infected WBC (vs in the cytosol)
Term
sx of ehrlichosis?
Definition
similar to other rickettsiae, but no rash. multi-system sx
Term
describe the cell wall of chlamydiae
Definition
modified gram-negative cell envelope. inner and outer cytoplasmic membrane, outer membrane with LPS, but with peptidoglycan replaced by a proteinaceous matrix
Term
describe chlamydiae's 2 stage developmental cycle
Definition
1. elementary body - non-growing, metabolically inert, relatively hardy extracellular stage. binds to variety of normal cell surface proteins that act as receptors for receptor-mediated endocytosis. after endocytosis --> EB prevents acidification of endocytic vesicle and fusion with lysosomes --> EB undergoes transition into reticulate body! 2. reticulate body - grows and divides like any other bacterial cell. form proteinaceous tubular structures that cross vesicle membrane and connect chlamydial and host cytoplasms, permitting direct import of host cell ATP and metabolites. hundreds of RBs accumulate, host cell deteriorates from depletion of nutritional supply, RBs develop into EBs that escape from the cell
Term
what are the 3 major chlamydial pathogens
Definition
chlamydia trachomatis, chlamydophilia pneumoniae, and chlamydophilia psittaci
Term
what are 3 conditions associated with chlamydia trachomatis
Definition
1. NGU (chlamydia) an exudative instead of ulcerative std 2. lymphogranuloma venereum - an ulcerative std assoc with massive lymphadenopathy 3. trachoma - chronic conjunctivitis, frequently leads to blindness
Term
what is the result of perinatal transmission of chlamydia?
Definition
baby gets pneumonia or conjunctivitis
Term
chlamydial NGU is the most frequent trigger for what?
Definition
reactive arthritis, an autoimmune sequelae
Term
what are the sx of chlamydophilia pneumoniae?
Definition
frequent cause of atypical pneumonia and URT inf. long incubation, dry cough, variably fever, confined to airways, no peripheral leukocytosis, infiltrates (minor, lower lobe, no lobar consolidation)
Term
what is the usual clinical presentation of chlamydophilia psittaci?
Definition
atypical pneumonia, from birds remember.
Term
what are 2 examples of diseases caused by legionella pneumophilia?
Definition
legionnaires disease and pontiac fever
Term
how are legionella diseases spread?
Definition
fastidious gamma proteobacterium, parasitizes several common aquatic amoebae and is acquired by inhalation or aspiration of water droplets containing them. outbreaks assoc with heating and cooling systems, more common in hot than cold water supplies, shower heads, supermarket produce misters(no person to person spread, just common source spread) common nosocomial infection
Term
how do legionellae get into cells and how do they multiply?
Definition
phagocytosed by macrophages using either C3 or Fc receptors --> products of their dot genes prevent phagosome-lysosome fusion --> multiply to high # within endosome --> escape as cell disintegrates
Term
where does the pathology in legionallae diseases come from?
Definition
results from inflammatory and immune responses to the infection - macrophages activated to kill or inhibit them via TH1 lymphocytes in immune individuals or to a lesser extent by TNF alpha produced in the inflammatory response in pre-immune individuals
Term
what are some clinical sx of legionellae diseases?
Definition
sx mimick pneumococcal pneumonia to some extent: lobar consolidation, alveolitis. nonspecific febrile influenza-like illness. point of distinction: frequent extrapulm sx affecting CNS (delirium, coma) or GI system (diarrhea).
Term
how do we diagnose legionallae disease?
Definition
best way: urine antigen detection. can plate on culture but must plate with charcoal to absorb growth inhibitors; can be visualized in sputum samples as faintly staining gram-negative coccobacilli
Term
what kind of drugs are used to tx legionallae?
Definition
drugs with good cell penetration i.e. macrolides, fluoroquinolones, tetracyclnes, sulfonamides, rifampin. not clinically responsive to beta-lactams bc poor cell penetration
Term
listeria monocytogenes, how do we get it? who is this most hazardous to?
Definition
acquired by eating uncooked food, esp refridgerated food like pate, milk, soft cheeses, deli meats, coleslaw. unusual capacity for growing at cold temps and resists moderate heating (ie pasteurization). most hazardous to immunocompromised adults and pregnant women
Term
how are listeria taken up by cells and how do they multiply/replicate?
Definition
complement-opsonized listeriae phagocytized in GI tract by macrophages via c3 receptors --> TH1 activated macrophages kill the bacteria, but in other cells bacteria lyse the primary phagosome using listeriolysin and phospholipase and escape into the cytoplasm --> multiply in cytoplasm, secrete ActA protein at one bacterial pole --> organize cellular actin into polar "tails" that provide them with intracellular motility and ability to push out filopoida in cytoplasmic membrane and break through into neighboring cells (rocket motility) --> initiate new round of intracellular multiplication
Term
what are the local and distant sx of listeriosis?
Definition
influenza-like sx (fever, myalgia), diarrhea. spread to CNS (meningitis, encephalitis, brain abscess) or joints and bones, lungs.
Term
what happens to pregnant women and their babies who are infected with listeria?
Definition
infection of placenta --> miscarriage or premature delivery or systemic inf of fetus w/disseminated abscesses or granulomas. perinatal inf --> neonatal meningitis. 3rd trimester is greatest risk bc increasing immunosuppression.
Term
how do we diagnose listeria monocytogenes?
Definition
readily gram stained and grows on routine media.
Term
how do we treat listeriosis?
Definition
gram +, so susceptible to beta-lactam antibiotics (pcn, etc)
Term
what is the organism that causes Q fever and what is the mechanism it uses to get into cells and how does it replicate once there?
Definition
Coxiella Burnetti. infects macrophages by phagocytosis, permits phagosome-lysosome fusion, persists in the acidified phagolysosome.
Term
how is coxiella burnetti spread?
Definition
zoonotic infection, primarily of livestock (sheep). high concentration in infected animal tissues, body fluids. is hardier and can be spread by inhalation or tick bites. largely an occupational disease, affecting farmers, vets, and lab workers.
Term
describe the 2 cell variants in coxiella burnetti development
Definition
large cell variant (actively dividing) and small cell variant (resting, hardy, accounts for its environmental persistance)
Term
describe the sx of q fever (c. burnetti)
Definition
multiple organ systems affected. elicits granulomatous host response. pathophysiology mostly immune-mediated. asymptomatic to influenza-like, self limiting. development of chronic infection (can manifest as endocarditis) is serious and results from granuloma formation and immune complex deposition
Term
how do we come into contact with brucellosis?
Definition
zoonosis.
Term
what is the intracellular mechanism of replication for brucellosis?
Definition
get phagocytized by macrophages, prevent phagosome-lysosome fusion, grow in phagosome, produce granulomatous reactions --> infected macrophages carry the pathogen to 2ndary lymphoid tissue and thence to all organ systems
Term
what are the 4 cardinal signs of acute inflammation?
Definition
redness (d/t congestion/vasodilation); warmth (d/t congestion/vasodilation); swelling (d/t edema/increased vascular permeability); pain (d/t chemicals? stretched nn?)
Term
what happens in the general response to injury? (when sublethal injury is serious enough)
Definition
mediators that are produced and act locally are in such great quantity (IL1, IL6, TNF alpha) --> into blood stream --> go to 1. liver --> induce liver to produce acute phase proteins (fibrinogen, c reactive protein, and serum amyloid protein); 2. go to the hypothalamus to thermal regulatory center to induce production of prostaglandin E2 --> sets thermic center temperature higher --> fever and 3. go to bone marrow to chase out sequestered neutrophils --> leukocytosis. also activate colony stimulating factors so leukocytes can be sustained
Term
what causes early permeability in inflammation?
Definition
histamine release (venules first to be affected. cause exudation as they dilate)
Term
what causes late permeability in inflammation?
Definition
clotting factor XII is activated --> activates PKA --> activates kalligreinogen --> cleaved into kallikrein --> made into kininogen --> made into kinins (bradykinin) --> late permeability (capillary damage, mediated by prostaglandin E)
Term
during inflammation, describe the process of neutrophils leaving the blood stream to travel to site of injury.
Definition
after stasis of laminar blood flow (caused by edema) --> neutrophils marginalize, adhere to selectins which are expressed both on the leukocyte and on the endothelium (only on endothelium when there's injury - IL1, IL6 and TNFa activate endothelium so these selectins are exposed) --> roll along selectins --> bind to integrins and become stuck --> migrate toward endothelial gaps --> neutrophils are then under the influence of chemoattractants
Term
how is necrosis brought about in foreign body caseating granulomas?
Definition
toxic irritant phagocytized --> toxic to macrophages, so rip up phagocytic membrane --> release enzymes into cytoplasm --> kill cell --> cell releases powerful enzymes into the environment --> necrosis
Term
how do phagocytes kill pathogens that are adapted to survive in the phagosome or lysosome?
Definition
1. everytime macrophage phagocytizes, undergoes respiratory burst --> rapid uptake of O2 and in presence of oxidase --> forms superoxide anion and H202 --> radical acts on organism, kills it. 2. myeloperoxidase - enzyme that, when in contact with H202 and a halide --> form H202-myeloperoxidase-halide-system --> hyperchloride/chloox = powerful antibac
Term
what is pus?
Definition
large # of dead and dying neutrophils, copious amounts of fibrin, necrotic debris, in an area of liquefactive necrosis (neutrophils get there, die, release enzymes, liquefy area)
Term
what happens when tissue cannot regenerate after acute inflammation (aka how do scars form)?
Definition
ingrowth of granulation tissue from the viable margins to replace the destroyed non-regenerating parenchyma. (starts in periphery and works its way into the abscess) mediated by vascular endothelial growth factors. when the vessels form, no tight junctions, so edema is common. granulation tissue = proliferating new capillaries and fibroblasts --> fibroblasts secrete collagen --> collagen shrinks and squeezes out capillaries and fibroblast nuclei --> left with avascular, acellular fibrosis/scar
Term
what are the causes of chronic inflammation?
Definition
when abscess does not heal properly, nondigestible persistant irritant (i.e. silica, TB), autoimmune rxn (where ag exits forever), persistent or repetitive injury, infection by resistant microbes
Term
macrophages are the distinguishing cells in chronic inflammation. what are their 2 main roles?
Definition
1. activated macrophages act as key scavenger cells, remove necrotic tissue 2. contribute to tissue destruction at sites of inflammation and stimulate repair and fibrosis through their release of growth factors and cytokines
Term
what is a granuloma?
Definition
focal, localized, circumscribed aggregate of macrophages or their transformed derivatives, the epithelioid cells, in solid tissue or highly confined space
Term
what is an immune granuloma and how do we get necrosis?
Definition
the antigenic components of the irritant (intracellular pathogen) evoke a vigorous cell-mediated IR. the immune (hypersensitivity) response --> local production of cytokines --> attract large numbers of macrophages and activates them --> upon dying these macrophages spill their cytokine-activated powerful lysosomal enzymes into surrounding tissues --> conspicuous necrosis
Term
what are non-caseating granulomas?
Definition
foreign substance inside that are not toxic nor antigenic. necrosis not seen. no collections of lymphocytes or plasma cells. granulomas are pure macrophagic granulomas.
Term
leukocyte rolling, what proteins/molecules are involved?
Definition
P and E selectins on endothelium, sialylated lewis X (oligosac) on neutrophil
Term
stable leukocyte adhesion, what proteins are involved?
Definition
ICAM-1 on endothelium, B2-integrin on neutrophil
Term
what 3 things mediate pain in inflammation?
Definition
bradykinin, PGE2, damaged nerves
Term
what 3 factors are important in scar formation?
Definition
TGF-B (most important), fibroblast growth factor, platelet derived growth factor
Term
what factors are involved in inflammatory vasodilation?
Definition
NO, PGI2, PGE2, platelet activating factor
Term
which genera of spirochetes has hooked tail?
Definition
leptospira
Term
which genera of spirochetes are anaerobic/facultatively anerobic/microerophilic?
Definition
treponema, borrelia
Term
which genera of spirochetes are aerobic?
Definition
leptospira
Term
how do all treponemas spread?
Definition
direct contact - are pathogenic only to humans, so cannot be cultured in vitro
Term
how are borrelias spread?
Definition
arthropod vectors (large, only ones that can be seen with LM)
Term
do syphilis infected individuals develop resistance to reinfection?
Definition
yes, but become susceptible again after treatment.
Term
what IR are active during syphilis?
Definition
TH1 response during primary and secondary syphilis, TH2 response predominates during resolution of lesions; both humoral and CMI necessary for protection
Term
describe the non-treponemal screening test in syphilis testing?
Definition
RPR - use phospholipid antigen, cardiolipin, which naturally occurs in genus treponema and mitochondiral membranes. are of prognostic value bc their titers decrease within weeks or revert to non-reactive in a few months of successful therapy
Term
what is the confirmatory treponemal test?
Definition
Fluorescent Treponemal Antibody - ABSorption (FTA-ABS). treat serum with treponema sp. extracts to remove nonspecific cross-reacting abs --> serum reacts with treponema pallidum on a microscope slide --> wash --> application of fluoresceien-labeled anti-human globulin
Term
what is the strain of spirochete that causes lyme disease? and what are the species of ticks that transmit it?
Definition
caused by borrelia burgdorfen; ixodes scapularis in eastern, I. pacificus in western. (reservoir: white footed mouse and white tailed deer)
Term
what organism causes relapsing fever? and what causes the fever to relapse?
Definition
borrelia hermsii, borrelia recurrentis; organisms undergo remarkable antigenic variation, relapses due to selection of variants with the major surface protein different from that which produced the previous relapse
Term
How do we contract Leptospirosis?
Definition
water contaminated with animal urine (worldwide disease of various mammals)
Term
what are the characteristic signs of the jarisch-herxheimer (J-H) reaction?
Definition
increase in temp and pulse, decrease in systolic and diastolic BP, increase in TNF-a IL-1 IL-6 and IL-8
Term
for weak acid: if pH of the medium is less than the pKa of the drug...
Definition
more unionized form will be present than ionized
Term
for a weak base: if pH of the medium is less than the pKa of the drug...
Definition
there will be more of the ionized form than the unionized
Term
what is bioavailability and what does it depend on?
Definition
fraction of administered drug reaching the systemic circulation. depends on fraction absorbed and first pass elimination. (measured as AUCoral/AUCiv
Term
what is bioequivalence and how do we measure it?
Definition
compares equivalence of two supposedly identical drugs. measure 1. area under curve (make sure both drugs are absorbed to the same extent) and 2. peak effect (make sure they have the same maximum concentration in the blood)
Term
how do you calculate apparent volume of distribution
Definition
Vd=apparent vol of distribution = dose/[drug] in plasma. [drug] extrapolated back to t=o on a [drug] vs. time plot and that concentration is plasma concentration
Term
how does infantile bilirubin poisoning happen?
Definition
infants have a high level of bilirubin (bc theyre hemolyzing RBC), which binds tightly to plasma albumin. infants have poor liver funcion and not a well formed BBB. Medications can displace bilirubin from albumin --> bilirubin diffuses into CNS --> toxic
Term
what protein binds to acidic drugs? basic drugs?
Definition
albumin binds to acidic drugs; alpha1 acid glycoprotein binds to basic drugs
Term
for urine, if you want to increase acidic drug excretion, what do you do to the urine pH and vv?
Definition
for increased acidic drug excretion, make pH more alkaline. for increased basic drug ecretion, make pH more acidic.
Term
how do resins and charcoals help us excrete drugs?
Definition
they absorb/bind to substances (drugs) by binding to them in the GI tract and preventing their absorption --> used to decrease their therapeutic effect
Term
drug metabolism: describe non-synthetic or phase I reaction
Definition
drug or metabolite undergoes hydrolysis, oxidation, or reduction
Term
drug metabolism: describe synthetic (conjugation) aka phase II reactions
Definition
drug or metabolite coupled to an endogenous substrate (carb, AA, glutathione, sulfate, etc) usually occurs after drug has gone thru a phase I reaction and requires energy
Term
the P450 enzymes associated with drug metabolism are associated mainly with what cellular organelle?
Definition
smooth ER
Term
in what organs are the P450s moreso associated with the mitochondria? what do they do there?
Definition
adrenal cortex, testes, ovaries (form pregnenolone from cholesterol) and in the adrenal cortex form cortisol from 11-deoxycortisol
Term
what is the general reaction of how drugs are metabolized by P450?
Definition
drug binds to oxidized P450(Fe3+) --> NADPH provides e- to reduce oxidized P450-drug complex (to Fe2+) --> molecular O2 associates with the complex, iron oxidized, oxycytochrome P450 complex formed --> NADPH provides another e- (sometimes NADH) and reduces oxycytochrome P450 complex forming active peroide anion species --> one atom of activated oxygen reduced to water --> second oxygen atom incorporated into substrate (drug) molecule --> oxidized P450 released
Term
what are 3 ways that P450 can be inhibited
Definition
1. competition of an endogenous compound or a drug for a particular species of P450 which metabolizes another drug. (thus, magnitude of inhibition tends to be dose related over the dosage range of the inhibitor) 2. inhibition as a result of destruction of the heme group of P450 as a result of the formation of a reactive metabolite of a drug during metabolism (ethinyl estradiol suicide substrate) 3. CO inhibits P450 activity by binding with the reduced drug-P450 complex
Term
what is therapeutic index? how does this affect the way we monitor P450 inhibition?
Definition
measure of the impunity with which you can give an overdose (if desired and lethal effects are far apart, wont have a problem, if they are close together, we must be very careful about P450 inhibition from other compounds/drugs)
Term
how does acetaminophen overdose work?
Definition
acitaminophen gets metabolized by P450 --> Nacetyl-p-benzoquinone imine --> binds to glutathione and eliminated as mercapturic acids (but if theres too much drug, GSH depletes --> covalent binding to nucleophilic centers in protein, dna, rna --> liver and kidney necrosis --> death)
Term
what does food deprivation do to the inducing effects of drugs on P450s?
Definition
enhances it
Term
what effect does feeding have on the inducing effects of drugs on the P450?
Definition
decreases
Term
describe how the process of glucuronidation leads to the excretion of endogenous compounds
Definition
UDP-glucuronyl transferase catalyzes the conjugation of UDP-glucuronic acid to seeral endogenous compounds --> glucuronides of most drugs (MW <300) are excreted in urine. higher compounds form glucuronides --> secreted into bile --> enter intestine --> compounds hydrolyzed by beta-glucuronidase --> reabsorbed --> again conjugated in liver --> excreted into bile
Term
what happens in glutathione conjugations?
Definition
glutathione S-transferases replace halo- nitro- sulfonate- or sulfonamide groups with GSH --> several additional rxns --> mercapturic acids (for excretion)
Term
in first order rxns: clearance is? and absolute rate of elimination is?
Definition
clearance is constant; absolute rate of elimination is not constant. V varies linearly with Cp (when Cp
Term
zero order rxn: clearance is? absolute rate of elimination is?
Definition
clearance is not constant; ARE is constant (bc removing the same AMOUNT of drug per unit time) V approaches Vmax (when Cp>Km) "saturation kinetics" bc our ability to eliminate is saturated
Term
describe time-dependent kinetics
Definition
when a drug is given --> you get an increase or decrease in regulation of metabolic enzymes in the liver --> then you get a change in the ability to eliminate drugs (bc of the very fact that the drug has been given)
Term
histamine functions as a neurotransmitter in the CNS to mediate what 3 things?
Definition
emesis center (triggers emesis), homeostasis (thirst, hungger), neuroendocrine function (ADH release)
Term
what are the 2 main pathways that histamine is metabolized through
Definition
imidazole-N-methyltransferase; diaminoxidase
Term
how do histamine liberators work?
Definition
directly stimulate histamine release from mast cells w/o prior sensitization (w/o IgE). raises intracellular calcium --> stimulates histamine release, causes mast cell lysis, or directly displaces histamine from inside mast cell
Term
which histamine receptors are responsible for vasodilation
Definition
H1 (faster onset, shorter duration); H2 (slower onset but longer duration)
Term
which histamine receptors are responsible for increased vascular permeability?
Definition
H1 (only receptor on endothelial cells) some of the plasma proteins that leak out are inflammatory themselves (c-reactive protein) and histamine is chemotatic for other inflammatory mediators
Term
what is the "triple response" observed after intradermal application of histamine
Definition
localized red spot (vasodilation of small vessels); flare (bright red flush 1cm beyond red spot; d/t axon mediated vasodilation via H1 receptors on peripheral nerve endings); wheal (local edema d/t increased vascular permeability H1)
Term
how does H1 mediate vasodilation?
Definition
located on vascular endothelial cells, leads to release of NO --> smooth muscle relaxation
Term
how do H2 receptors mediate vasodilation?
Definition
located on vascular smooth muscle. rise in cAMP and decrease in intracellular Ca --> Ca is a stimulator of mm. contraction, so decrease in Ca --> relaxation of vessel
Term
how does increase in histamine cause bronchoconstriction?
Definition
H1 receptors on bronchial smooth muscle, constrictor effect mediated by increased IP3 --> rise in IC Ca --> bronchoconstriction
Term
which histamine receptors are important in mediating emesis, especially motion sickness?
Definition
H1
Term
which histamine receptors mediate homeostasis?
Definition
H1 and H2
Term
where are histamine receptors located in the stomach and what do they do?
Definition
on parietal (HCl-secreting cells)cells. mediate release of gastric acid and pepsin
Term
are 1st generation or 2nd generation agents more useful in combating bronchospasm?
Definition
2nd generation bc they may additionally inhibit mediator release from mast cells and basophils (whereas 1st generation does not usually block completely bc other mediators are usually involved)
Term
how do H1-antagonists exert local anesthetic actions?
Definition
via blockade of sodium channels on nerve cells
Term
why shouldnt patients with glaucoma take 1st generation H1 blockers?
Definition
1st gen drugs have a cros-over specificity with other receptor types (cholinergic) --> drugs can have anti-cholinergic function that manifests as anti-muscarinic effects (drying of nose and mouth, blurred vision)
Term
explain the significance of COXI and COXII
Definition
arachidonic acid found in biological membranes, esterified into phospholipids. phospholipase A2 de-esterifies arachidonic acid and bc it is unstable, arachidonic acid will be rapidly metabolized into eicosinoids via cyclooxygenase, lipoxygenase, and cytochrome P450. COX I and COX II are the 2 forms of cyclooxygenase - COX I constitutively present, involved in maintaining gastric cytoprotection. COX II is an immediate early response gene product that is induced 10fold by cytokines, GF, and tumor promoters (form prostaglandins)
Term
the lipoxygenase pathway of arachidonic acid metabolism yields what?
Definition
most commonly: 5-lipoxygenase (in white cells), 12-lipoxygenase (in platelets), and 15-lipoxygenase (in macrophages and epithelial cells). 5-lipoxygenase --> leukotrienes
Term
what does 5-lipoxygenase produce?
Definition
hydroperoxide (5-HPETE) --> LTA4 --> LTB4 (leukocyte chemoattractant) or LTC4/LTD4/LTE4 (peptidoleukotrienes that form the slow-reacting substance of anaphylaxis, likely the physiological factors that cause bronchospasm)
Term
what is the receptor for LTB4 and where is it located?
Definition
BLT - neutrophils, lung, spleen
Term
where is the receptor for LTD4 and LTE4 located and what is it?
Definition
cysLT1 - smooth muscle, especially in lung
Term
what is the receptor for LTC4 and where is it located?
Definition
cysLT2 - endothelium and smooth muscle
Term
what determines which prostaglandins are formed in cells?
Definition
from arachidonic acid, COX II makes PGG2 --> gets converted by many diff enzymes into various other prostaglandins depending on what enzyme is present in which cell (lung and spleen produce most products; platelets only have thromboxane synthase; endothelial cells only have prostacyclin synthase)
Term
the adenylate cyclase pathway (AC) vs calcium calmodulin pathway (PLC) - how do we know which causes constriction and which causes dilation?
Definition
Myosin light chain gets phosphorylated by ATP. as rxn moves in direction of MLC phosphorylation (PLC pathway) we get constriction; as rxn moves in direction of dephosphorylation (AC pathway) we get relaxation/dilation
Term
which prostaglandins generally go thru the AC route (relaxation)?
Definition
PGI (prostacyclin) PGA and sometimes PGE
Term
which prostaglandins go thru the PLC (contraction) route?
Definition
PGF, TXA2, PGH2, leukotrienes
Term
compare CBG and albumin
Definition
CBG is a high affinity low capacity binder of glucocorticoids. albumin is a low affinity high capacity binder
Term
why is 11-betahydroxysteroid important?
Definition
inactive cortisone cannot bind to mineralcorticoid OR glucocorticoid receptors (must be converted to active cortisol by 11-beta-hydroxysteroid 1 in order to bind to glucocorticoid and mineralcorticoid receptors) cortisol binds the mineralcorticoid receptor for aldosterone with high affinity, so in order to keep the MR from being swamped by cortisol, 11-beta-hydroxysteroid 2 converts it to inactive cortisone.
Term
how do glucocorticoids affect carbohydrate and protein metabolism?
Definition
decrease glucose uptake and utilization; also mobilize AA which serve as substrates for hepatic glycogen and glucose production (activities of several heptaic enzymes involved in gluconeogenesis enhanced). results in tendency twds hyperglycemia
Term
what do high doses of glucocorticoids do to muscle?
Definition
causes muscle wasting. steroid-induced myopathy can result from prolonged glucocorticoid therapy
Term
what is the effect of glucocorticoids on cell growth and division?
Definition
can inhibit cell division or DNA synthesis (reduce growth in children, premature closure of epiphyseal plates, delay wound healing, skin thinning, GI ulcerations)
Term
what effect do glucocorticoids have on prostaglandin synthesis?
Definition
inhibit COXII and induce lipocortin (phospholipid binding protein) which reduces the availability of the phospholipid substrates of phospholipases (inhibits release of arachidonic acid), inhibits phospholipase A2
Term
what is NF-kB and how does glucocorticoid interfere with it?
Definition
NF-kB is an essential transcription factor for a variety of pro-inflammatory cytokines, inflammatory enzymes, adhesion molecules, chemokynes, receptors. NF-kB sequestered in cytoplasm by I-kB proteins. upon activation by inflammatory stimuli, I-kB degraded and NF-kB enters nucleus and transcribes products for inflammation. glucocorticoids induce synthesis of I-kBa which binds to and inhibits action of NF-kB
Term
what are the 3 major ways that glucocorticoids influence immune system?
Definition
1. redistribute circulating leukocytes (causing neutrophilia, lymphocytopenia, monocytopenia, basopenia, eosinopenia) 2. inhibits T-cell activation and proliferation 3. inhibits cytokine synthesis by interfering with transcription
Supporting users have an ad free experience!