• 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

1/1264

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;

1264 Cards in this Set

  • Front
  • Back
Roles of hormones
"Reproduction, growth, homeostasis, EPU"
Alpha Cells Secrete
Glucagon
Beta Cells secrete
insulin
insulin begins as
preproinsulin
First step of processing of preproinsulin
"Cleaving off the signal peptide, becomes proinsulin"
where is proinsulin processed
Golgi
Why is the C-peptide important
can monitor amount of insulin secretion. made in 1:1 ratio. cleaved of A and B chains to make proinsulin into insulin
where is preproinsulin synthesized
cytosol
what is one of the primary regulators of insulin secretion
ATP/ADP levels in beta cells open/close K channels which then effect Ca channels which release the vesicles
What cells are affected by insulin
Only cells that have insulin receptors
what glucose transporters are in pancreatic B cells
GLUT2
What is the primary effect of insulin
Enhances translocation of GLUT4 to the membrane of target cell
In general what physiologic processes does insulin produce
anabolic paths for storage with decreased catabolism
Glucagon is inhibited by
Glucose and insulin
how to catecholamines and glucocorticoids affect glucagon secretion
counteract insulin affects
What cell signalling pathway interacts with glucagon secretion
G protein coupling with PKA and cAMP
Whats up with glucagon in type I diabetes
since no insulin is produced glucagon is not inhibitted
What Glucose transporters have the lowest Km
1 and 3
What is the state of phosphorilated PFK2 in the liver
Inactive
"What is the state of Fructose 2,6 Bisphosphotase when there is no phosphorylation in the muscle"
Active
What is the result of glucocorticoids on a cell
"regulate transcription, resulting in slow changes that can have prolonged effects"
Leptin has what effect
satiey
Orexigenic or Anorexigenic: Gherlin
Orexigenic
Orexigenic or Anorexigenic: PYY
Anorexigenic
gamma bond of ATP releases how much energy
"7.3 kcal
"why is 1,3 biphosphoglycerate important"
"substrate level phosphorylation
When is substrate level phosphorylation especially important
RBC since they do not have a TCA cycle
What is the equation for BMR
Weight (kg) x 24 kcal/kg/day
TEE?
Total Energy Expenditure=BMR+activity
What is the go to fuel for muscle
Fatty acids
RBCs use what for fuel
ONLY glucose
What can't the brain use for fuel
Fatty acids
What is the livers role in metabolism
"Maintains all the other organs, produces KB, converts glucose to fat or glycogen for storage"
Where are fuels going in the fed state
"brain and rbc are using glucose for energy. adipose and liver are storing glucose, muscle is using glucose and other tissues are using proteins"
what shift happens in fasting
"production of ketone bodies, muscle is using more FA and KB, adipose is releasing FA and liver is going thru GNG to feed RBC and brain. Urea cycle kicks in"
Starved state has what effects
Urea cycle stops and brain is using KB exclusively. All glucose made from GNG is going to RBCs
how many protons are pumped by oxidizing NADH
"10, 4 from I, 2 from III, and 4 from IV"
How many protons are pumped by oxidizing FADH
"6, 2 from III and 4 from IV"
F0 of ATP synthase is located where
imbedded in the inner membrane of mitochondria
what subunit is turning in ATP synthase during proton transport
Gamma
"in one full turn of gamma, how many protons are pulled thru, and how many ATP are made"
12 H+ and 3 ATP
How does ADP get into the matrix
"Use of an antiporter with ATP
how is P being replenished in the matrix
A symporter Phosphate translocase moves H2PO4 and H+
how do reducing equivalents get into the mitochondria
Malate Aspartate shuttle
Malate Dehydrogenase
OAA+NADH-->malate
Aspartate Aminotransferase
"Transamination of OAA to Asp
What does the brain do to get reducing equivalents
"Glycerol 3 phosphate shuttle
What is Oligomycin
"blocks proton channel of atp synthase. enhances ETC at same time until proton pumps fail, slows overall o2 consumption"
what is DNP (dinitrophenol)
disconnects proton pump from ETC. Disrupts proton gradient. ADP builds up and forces ETC but no ATP made
Thermogenin
uncouples proton gradient produces heat
what enantomer form are most monosacharides in
D-form
Which configuration is glucose most commonly found in
Alpha. OH on C1 is opposite C6
Galactose+ Glucose=
Lactose
Fructose+ Glucose=
Sucrose
Dietary Fiber has what type of glycosidic bond
"B 1,4"
Glut5
Fructose transporter
Glut1
glucose and galactose
SGLT1
Na and Glucose cotransporter
how does a cell reestablish Na concentration in regards to SGLT1
Na K ATPase pump
What is the net production of glycolysis
2 ATP and 2 NADH (.5 ATP thru TCA)
Hexokinase is inhibitted by
Glucose 6 Phosphate
Glucokinase is inhibitted by
Fructose 6 Phosphate
What is positive feedback for PFKII
"Insulin, Ca"
What pushes F26Bp towards F16Bp
Glucagon
What inhibits PFKI
"ATP, Citrate"
What induces PFKI
"AMP, F2,6Bp"
What is the rate limiting step of glycolysis
PFKI
Inhibitors of PK (pyruvate kinase)
"Alanine, ATP"
Activator of PK
"F1,6P (insulin in the liver)"
At what steps of glycolysis is ATP produced
"phosphoglycerate Kinase, pyruvate kinase"
What steps of glycolysis require energy
"Hexokinase/glucokinase, PFK"
What can pyruvate become
"Alanine, lactate, OAA, acetyl CoA"
when is NADH produced in glycolysis
"Glyceraldehyde 3 phosphate--> 1,3 bisphosphoglycerate"
How is Glucokinase regulated
"sequestered in nucleus with a regulatory protein, glucose dissociates the RP. F6P activates the RP"
1 Trip thu TCA gives
"3 NADH, FADH, GTP"
What is the driving force of TCA
"Low NADH/NAD+ ratio, Low ATP/ADP"
What enzymes are regulated in TCA
"citrate synthase, isocitrate dehydro, a-ketodehydeo, succinate dehydro"
where does the pyruvate dehydro rxn happen
mitochondria
What moves pyruvate into the mitochondria
monocarboxylate transporter (antiporter with OH)
PDH complex has what vitamin as a part of it
"Thiamine (B1), B2, and B3"
What regulates PDH
"substrate activation, product inhibition, PDH kinase/phosphotase, kinase and phos are further regulated by products and substrates of PDH"
PDH deficiency
Leads to Lactic acidosis
what is the substrate of the PPP
G6P is converted to a pentose sugar while producing NADPH via oxidative path or F6P via the non oxidative path
What glucosylates glycogenin
itself!
insulin does what to glycogen synthesis enzymes
"activates the phosphotases, no phosphorylated enzymes= glycogen synthesis"
What enzyme is not present in muscle regarding glycogen
Glucose 6 phosphatase. that way g6p can only go thru glycolysis
UDP Glucose Pyrophosphorylase
makes G1P into UDP Glucose
Glycogen Synthase
"creates 1,4 glycogen bonds when not phosphorylated"
4:6 Transferase
makes 1:6 glycogen bonds to create branching in glycogen after about 11 glucoses are added in a chain
Why does glycogen need to branch
so there are many available reducing ends to add and subtract glucose molecules from
When glucogon is high what regulatory enzyme is turned on regarding glycogen
Kinase. when glycogen enzymes are phosphorylated glycogen breakdown predominates
what interconverts G6P and G1P
phosphoglucomutatse
"4,4 transferase"
"takes off branches 3 Gs from branch point and creates 1,4 bonds"
"what is special about the action of 1,6 glucosidase"
"removes the branch point as glucose, not G6P"
What is the interaction between PK and epi in muscle tissue
Epi will not cause phosphorylation of PK
What is the deficiency in McArdle's Disease
Glycogen phosphorylase
what 2 chromosomes are most hemoglobin genes on
11 and 16
What 2 types of hemoglobin are hb gower
epsilon and zeta
Hb F is which 2 globins
alpha and gamma
what is HbA2
alpha and delta. low abundance adult hb
"What Hb has the highest affinity for O2, why?"
"HbF, so it can steal O2 from mothers blood stream"
What creates methemoglobin
oxidizing iron to become Fe3+
F helix is proximal or distal
Proximal Histadine
what does the distal histidine do
stabilizes the O2 interaction and prevents oxidation and reduces affinity for CO
What increase Fe absorbtion
Vit C
What breaks down iron compounds after theyve been absorbed
heme oxygenase. to be absorbed Fe3+ in diet must be reduced. reductase upregulated by Vit C
What must happen to iron to be carried by transferrin
transferrin only carries Fe3+ so it must be oxidized again by ferroxidase
"If a pt has a high TIBC, what does that mean"
the pt is iron deficient
What are agents that inhibit heme production
"Heme, Heavy Metals"
What would happen in a ferrochetalase deficiency
"heme would still bind Fe, but at much slower rates"
Which step in heme synthesis is the most sensitive to enzyme dysfunction
"Step 4, takes the unstable compounds and turns them into heme, without enzyme 4 many unstable dangerous metabolites can be formed"
What is the difference between ALAS1 and 2
"ALAS1 makes heme for tissues other than blood, ALAS2 makes heme for blood"
How do cells ensure there is no disproportionate levels of heme to globin
globin synthesis is induced by presence of heme and inhibitted thru HCI if no heme is available
What would be a good way to treat a pt with toxic levels of heme metabolites
inhibit ALA synthase (hematin or glucose) which will slow the production at the rate limiting step
Porphyria Cutanea Tarda
"Dom inher.
Acute Intermittent Porphyria
colored stool and urine. agitation/madness. mis-Dx as psych issue
what contributes to heme being able to release O2 to tissues
"physical strain on molecule, likes to be puckered but O2 flattens it out."
which steps of heme synthesis happen in the mitochondria
1st and last 3
2.3 BPG levels _____ in people living in higher altitudes
Higher
"explain why 2,3 BPG levels vary in people of different altitudes"
"2,3 BPG decreases hb affinity for O2, allowing more effective O2 delivery to peripheral tissues."
"besides altitude what can increase 2,3 BPG levels"
"Anemia, Cardiopulmonary insufficiency"
Explain the Bohr effect
"as O2 binds to hb, H+ is released. If the blood becomes acidotic it pushes O2 off hb, decreasing affinity."
alpha thalassemia
"deletion of 1 or more alpha globin genes. 1 deletion-silent. hydops fetalis, all 4 deleted, hemoglobin H, 3 deletions"
B-thalassemia
"varied phenotype. minor-severe. in severe cases too much a-globin, forms insoluable aggregates."
what is the regulated step of Hb degradation
"heme oxygenase cleaves pyrrole ring, requires O2 and NADPH. Releases CO and Fe3+. product: biliverdin"
what can physiologic levels of CO tell a clinician
Rate of heme breakdown
what are the steps of bilirubin conjugation
"Bili is carried to liver on albumin
what moves conjugated bili out of hepatocytes
multidrug resistance protien 2
what does direct bilirubin measure
conjugated bilirubin
What is a characteristic of prehepatic jaundice
"no bili in urine, increased unconjugated bili"
intrahepatic jaundice will result in what lab results
increased urobilinogen in urine and decreased in stool.
Posthepatic jaundice presents:
"lots of conjugated bili in urine, no urobili in stool or urine"
What does Haptoglobin do
binds loose ab hb dimers in blood
"What are the caloric values of carbs, protein, fat, and ethanol"
"4, 4, 9, 7"
what is the relationship between length of FA and melting point
longer chains make for higher melting points
how do double bonds affect melting point
"more double bonds, lower melting point"
ARDS is a result of:
lack of phosphatidylcholine (lecithin)
What vitamin is important for myelin
B12
the major component of cholesterol is
Acetyl CoA
How do Lipase and Colipase work together
colipase makes space for lipase to work in the micele to make monoacylglycerol
what happens to MAG and FA in absorptive cells in the intestines
reassembled as TG and packaged into chylomicrons and sent to the lymphatics
Small and medium chain fatty acids are special why
Don't need micelles or chylomicrons. long and very long do
which lipoprotein is located on chylomicrons
ApoB48
HDL gives up which proteins to chylomicrons
ApoCII and ApoE
How do chylomicrons get into cells
LPL is upregulated by insulin and binds to ApoCII in the capillary lumen. digests TG in the chylomicron to FA and glycerol
Describe the actions of hormone sensitive lipase
"Epi, Glucagon, corticoids activate PKA which P-ates HSL. HSL removes FA from glycerol 1 by 1"
how do LC and VLC FA get into the mitochondria
"LC are P-ated and bound to CoA, carnitine shuttle moves it in. VLC are reduced to LC in peroxisomes"
Describe the process in which LCFA gets P-ated
ATP releases 2P and binds AMP to LFCA via Thiokinase. CoA is added and AMP is released
How does the Carnitine Transport System work
CPTI on outer membrane swaps the CoA for carnitine. FA-Carn then enters antiporter carnitine acyl carnitine translocase into the matrix. CPTII on inner membrane swaps carnitine for CoA
What inhibits CPTI
Malonyl CoA (FA synthesis intermediate)
What is produced from each round of B-oxidation
"1 NADH, 1 FADH2, 1 Acetyl CoA"
If there is an error in MCFA dehydrogenase what will happen
B-ox cannot happen and will result in low ketones and eventually liver damage from ammonia build up
What happens if a FA has an odd # of carbons
"get propionyl CoA, eventually becomes Succinyl CoA. Uses biotin and B12"
Gaucher's Disease
Marcophages cant break down glucocerebroside. get accumulation
What is the first barrier in GNG
Pyruvate-OAA-PEP
What are the components of the 1st regulated step of GNG
"Pyruvate+Biotin,CO2, ATP, Pyruvate Carboxylase. PCarb is allosteric by acetyl CoA"
What is the total energy input for the first step of GNG
"1 ATP, 1 GTP for each Pyruvate-->PEP"
How does B oxidation tie into GNG
"lots of acyl CoA around, will allosterically activate rxn Pyruvate -->OAA"
what enzyme catalyzes the reverse reaction of hexokinase
Glucose 6 Phosphatase
What is the GNG equivalent of PFKI
"Fructose 1,6 Bisphosphatase"
How are ketone bodies formed
"2 acetyl CoA attach, HMG CoA in mitochondria adds another acetyl CoA. lose an acetyl CoA, get acetoacetate as an end product."
how do you get B hydroxybuterate
oxidize acetoacetate with NADH.
how do ketone bodies make energy
"from BHB, creates NADH, succinyl CoA and 2 Acetyl CoA for TCA. Acetoacetate creates succinyl CoA and 2 Acetyl CoA"
Apo A1
"On HDL Receptor ligand for liver, activates PCAT"
B100
"on LDL, VLDL."
B48
On chylomicrons
CII
Activates LPL
CIII
inhibits LPL and Apo E
Apo E
Ligand for liver receptor
How does the protein transfer between Chylo and HDL occur
"HDL give Apo E and Apo CII to chylo. after chylo gives up TG, it gives Apo CII back to HDL. Keeps Apo E so it can dock at the liver"
Describe the path of cholesterol from VLDL
VLDL has Apo B100. Gets E and CII from HDL. GIves off TG as CII binds LPL. Becomes LDL. LDL gives back CII and E to HDL. B100 signals to tissues to uptake the LDL.
What is an ACAT reaction
makes cholesterol Esters
What is HDLs path
starts as A1 with some fat and interacts with ABCA. Takes on cholesterol. Pancake stage. PCAT begins making cholesterol esters. rounds up. brings these back to the liver.
how does HDL interact with VLDL besides protein exchange
VLDL gives HDL phospholipids in exchange for TG. PCAT is taking phospholipids and making esters. so HDL needs more PL.
What is evidence of Metabolic Syndrome
"Pear shaped, High TG, Low HDL, HTN, Hyperglycemia, Albuminuria"
What are the risk factors for CHD
"Age, Fam Hx, Smoking, HTN, Low HDL, Diabetes, Obesity"
3 things you need for FA synthesis are
"Acetyl CoA, Energy, Reducing power (NADPH)"
What is the first step of FA synthesis
Convert Acetyl CoA to Malonyl CoA
How can the body prevent wasting of materials and energy during FA Syn
while transferring reducing eq to NADPH pyruvate is created. OAA->Malate->pyruvate
Where is the most regulation in FA Syn
"Acetyl CoA->Malonyl CoA
what vitamin is essential for Acyl Carrier Protein
B5
What happens in the priming step of FA syn
Acetyl group attaches to the SH and then is transferred to the cysteine nub
What occurs in the first reaction of FA syn
"Malonyl attaches to the SH and in a condensation reaction bonds to the acyl group on the cys nub, CO2 is released."
to go through one cycle of FA Syn what is consumed and produced
2 NADPH are used. 1 CO2 and 1 H2O are produced
What is a restriction of FA synthase
Can only make FA 18 carbons or shorter
Where is FA elongase located
On the ER
How do you make double bonds on new FA
FA CoA desaturase. uses O2. makes double bond and oxidizes NADH.
what is FA CoA desaturase limitation
can only make double bonds up to carbon 9
What regulates Acetyl CoA Carboxy
Citrate activates and palmitoyl CoA inhibits. Phosphorylation by glucagon will inhibit.
How do we prevent a futile cycle of FA Syn and B-oxi
"synthesis happens in cytosol, b-ox in the mitochondria. malonyl CoA also prevents b-oxi substrates from entering mitochondria"
what enzyme is lacking in adipose to prevent futile cycling of TG syn and breakdown
no glycerol kinase to break them down.
what illnesses are related to diet
"scurvy, goiter, rickets, diabetes, CHD, osteoarthritis, osteoporosis"
Some Fat soluable Vitamins
A D E K
What are the most important D vitamins
Ergocalciferol (D2) and cholecalciferol (D3)
Vitamin D deficiency=
Osteomalacia
What is Vit D's job
regulates Ca via absorbtion
What is released by parathyroid glands and what does it to to regulate Ca
"PTH, stimulates osteoclasts"
Vitamin K function
"Coagulation Proteins, bone mineralization"
Sources of Vit K
"Green leafy veg, intestinal bacteria"
Vit K deficiency
"very rare, clotting delays. megadosing of A and E can counteract K"
What does thiamine do
"B1 is par of TPP, removes carboxyl groups in glucose metab. Used in B-oxi and nervous system"
Thiamine Deficiency
"BeriBeri-muscle weakness, heart failure. Wet beriberi has edema. ETOH associated"
Riboflavin
"B2. part of FADH, in TCA and B-oxi. aids in deamination"
Riboflavin Deficiency
Angular Stomatitis-cracks in the mouth
Sources of Riboflavin
"Liver and yogurt, some milk"
Niacin
can lower LDL via HMG reductase inhibition. Part of NADH.
Sources of Niacin
"Tryptophan, meat, grains (minus corn)"
Deficiencies of Niacin
"Pellegra. the 4Ds dementia, diarrhea, dermatitis, Death"
Pantothenic Acid
B5. part of CoA
Sources of Pantothenic acid
"liver, in lots of food"
Pantothenic Acid Deficiency
"None, unless induced"
Biotin
Can be made in the bowel by bacteria. used in carboxylation rxns.
Biotin Deficiencies
"Leiners disease-seborrheic dermatitis, baldness, waxy skin."
B6
"Three forms. Pyridoxal, pyridoxine, pyridoxamine."
B6 function
transamination. blood cell synthesis. influences homocysteine levels
Deficiency of B6
"Microcytic hypochromic anemia, atherosclerosis"
Folate
"Converted to THFA, used in one carbon metabolism in DNA syn"
Folate deficiency
"megaloblastic anemia, spina bifida"
B12
works with folate. mylelin sheath. works with TCA
What is the difference between major minerals and trace elements
"Major minerals are needed in concentrations >100mg, trace elements <100 mg"
What is the most key aspect to mineral absorption
Stomach acidity keeps the minerals ionized and soluable
What causes Na deficiency
"disease state (vomiting, diarrhea, sweating)"
What is are some comorbitities with hyponatremia
"dehydration, edema, neurological issues"
Functions of Na
"fluid balance, nerve transmission, muscle contraction, blood pH, nutrient absorption"
What occurs in hypernatremia
hypervolemia-->HTN
What can loop diuretics decrease
"K, Mg levels"
Hypokalemia
"Abnormal cardiac rhythm, muscle cramps"
Hyperkalemia
"Rare, except in cases of renal disease. Heart arrhythmias"
Hypocholoremia
can result in metabolic alkalosis.
Hypophosphotemia
elevated PTH. Vit D deficiency. results in osteomalacia
Hyperphosphotemia
"kidney disease, low PTH level. too much Vit D. "
What should be a concern when P is high and Ca is low
Excessive bone loss
Hypomagnesaemia
"bleeding problems, stiffness, cramps"
Zinc Deficiencies
"Hypogonadism, hair loss, skin lesions, growth retardation, heart disease"
Selenium
part of glutathione peroxidase
Chromium
involved in homeostasis of glucose levels
what are the five factors determining state of nutrition
"biological, social, psychological, presence of disease, energy expenditure"
What does the EAR represent
dietary intake that will meet 50% of the requirement at their particular stage of life
what is the RDA
the dietary intake that will meet 97-98% of dietary requirements
what are the drug classes that sensitize the body to insulin
bigaunides and thiazolidinediones
what drug classes increase insulin secretion
"Sulfonylureas, meglitinides"
What do alpha-glucocosidase inhibitors do
"slow the absorbtion of starches, create alot of gas"
what is byetta
"gila monster saliva, curbs appetite, affects leptin"
januvia
another diabetes drug
What is the difference between Gprotein coupled receptors and enzyme coupled receptors
GPCRs act indirectly and send a signal molecule to the enzyme. ECR have either enzymatic activity on receptor or right next to the receptor
what are the 3 small 2ndary messagers
"DAG-lipid sol, Ca and cAMP-water sol"
what are the 8 mechanisms of large 2ndary messengers
"scaffold, transform, amplify, relay, spread, anchor, modulate, integrate"
What are the 3 amino groups that usually get Pated
"serine/threonine, tyrosine"
What do SH2 domains bind
P-ated tyrosine
what do SH3 domains bind
non P-ated proline rich areas
What does PTB bind
P-ated Tyrosine
What do Pleckstrin Homology domains bind
phosphoinositides
Explain the inositol phosphilipid signal path
"PLCB activated
What is PKC
Ser/Thr kinase
What is Calmodulin
"Ca Carrier. binding causes conformation changes, can influence transcription factors"
What are the 4 major classes of adhesion molecules
"selectins, intergrins, ICAMs, cadherins"
What is the most important class of cell-cell molecules
Cadherins
What mediates the strength of cadherin adhesions
number of cadherin molecules (like velcro)
What does the intracellular domain of a cadherin do
"initiates signalling, creates scaffolds"
what does the extracellular domain of cadherins do
"binds ligands, receptor for adhesion molecules"
What happens to cadherin molecules in the absence of Ca
the wobble and lose their adhesion to other molecules
Besides adhesion what is another role of cadherins
"regulate space between cells, anchor cytoskeleton elements"
what molecule is important to cadherin attachment to the cytoskeleton
B-catenin
If there is a gene mutation in B catenin:
easy disaggregation resulting in local invasion and distant metastasis
What cancers are closely related to cadherin disfunction
"breast, esophagus, colon, ovary, prostate"
What is the other function of B-catenin
gene regulatory protein signalling for cell replication
in colon cancer what happens in B-catenin signalling
cell believes that WNT is continuously signalling and B-catenin is not destroyed therefore promoting call replication
What is the primary ligand for selectins
carbohydrates
what adhesion molecules are Ca dependent
cadherins and selectins
What are the functions of selectins
"homing receptors for lymphocytes to enter lymph nodes, facilitates binding of neutrophils to endothelial cells (rolling interactions)"
what 3 endothelial ligands bind L-selectin
"glycam, madcam, CD34"
What do E selectins bind (ELAM)
"Lewis A or Lewis X family molecules on granulocytes, monocytes, or memory T cells"
Why is E selectin important
brings effector lymphocytes to sites of inflammation
Where is P selectin stored in platelets
Wiebel-Palade bodies
Why are p selectins stored
because platelets cannot synthesize proteins
What disorder in humans is related to selectins
Leukocyte adhesion deficiency 2. susceptible to bacteria infections
What do integrins do
coordinate and integrate extracellular signals with motility and shape changes of cell
What is the structure of integrins
Heterodimers with alpha and beta chains. types are named by the beta chain.
"extracellularly, what to integrins bind"
RGD in fibronectin especially
what is the trick to recognizing a cytoskeletal component that binds integrins
"ends with IN (talIN, actIN, tropomyosIN, actinIN, vinculIN)"
What is another name for B1 integrins
VLA (very late activating) also CD49 (a-g subtypes)
where are VLAs expressed
"constitutive on leukocytes, can be induced elsewhere"
What is the primary function of B1 selectins
homing of lymphocytes to areas of inflammation
What is the other name for B2 integrins
"LFA- lymphocyte function associated antigen
LFA1 has the CD#:
CD11aCD18
what does LFA1 bind
ICAM on APCs
What is a deficiency in B2 integrins
"LAD1 no B2 integrins, succumb to bacterial infection"
What are hallmarks of apoptosis
"condensed chromatin, no inflammation, distinct organelles, blebs. cell shrinkage, multiple nuclei, "
Biochemically what happens in apoptosis
"Its energy dependent, caspases cleave proteins (lamins, scaffolds, cytoskeleton), dna breakdown, phosphatidyl serines flip for phagocytic recognition."
What induces apoptosis intrinsiclly
mitochondrial walls weaken and release cytochrome C.
What are Bcl-2 and Bcl-x
anti-apoptotic molecules on the mitochondrial membrane. inhibit apaf-1 and prevent pore formation on mitochondria
what are the pro-apoptotic members of the bcl2 family
"Bax, Bim, Bak"
How does cytochrome C induce apoptosis
activates caspase 9 by forming a complex with apaf1
How is extrinsic apoptosis intiated?
Fas ligand binds to a Fas receptor (TNF superfamily)
How does the Fas pathway work
"3 or more Fas molecules become linked by Fas ligand, FADD binds pro-caspase-8, pro-caspase-8's cleave each other and waterfall occurs"
What can inhibit Fas pathway
FLIP
How does the TNF-R path work?
Same as Fas but uses TRADD forms and binds FADD and so forth.
What are the main initiator caspases
8 and 9
What are the main executioner caspases
3 and 4
What does caspase 3 directly activate
"endonucleases, split DNA into large chunks (300 kbp) then 180 bp"
p53 has what role in apoptosis
"arrests cell cycle after G1 if DNA is damaged, if not repaired, p53 signals for apoptosis"
What is morphology as it relates to pathology
"What a cell looks like- shrink, swell, disintegrate"
What factors are important in determining reversible vs irreversible damage
"how long the damage is occuring, how severe is the damage, how hardy is the cell"
what are ways cells and tissues adapt to an injurious stimuli
"atrophy, hypertrophy, hyperplasia, metaplasia"
What is hypertrophy
"increased CELL size, stress on cell makes it work harder (muscle tissue)"
What happens in sub cellular hypertrophy
"organelles grow in size to meet demands. ER- increased detox, increased protein load. Mitochondria- increased energy demand"
What is hyperplasia
"Increased # of cells, usually in response to hormone changes (endometrium)"
why would hyperplasia occur physiologically
in response to hormones or damage to a tissue (remove part of the liver)
Atrophy is
Decreased cell size
when is atrophy physiologic
during development of embryo
pathologic atrophy can be caused by
"decreased use, blood supply, nutrition, nerve stimulation, or increased physical pressure on a cell"
metaplasia is
a hardier cell type replaces another sensitive cell type (columnar -> squamous)
what are indicators of necrosis
"cellular swelling, swelling and disintegration of organelles, "
What are some mechanisms of cell injury
"depletion of ATP, mitochondrial damage, influx of Ca, ROS, membrane permeability"
What occurs during ATP depletion
"decreased Na pump activity, water remains in cell and it swells. decreases pH thru glycolysis, cant pump Ca out, cant synthesize proteins for functioning."
Why does mitochondrial damage injure cells
"release cytochrome C leading to apoptosis, and decreases cell ATP."
How does Ca homeostasis injure a cell
"Ca accumulates in ER and mitochondria, damage membranes, cell explodes"
Necrosis displays what core signs
"membrane integrity is lost, inflammation, proteins are broken down and destroyed, necrotic cells are removed."
What is the morphology of necrosis
"eosiniphilic, myelin figures, nuclear changes"
what is karyolysis
nucleus broken down into fine fragments
what is pyknosis
nucleus shriveling up
what is karyorrhexis
nucleus breaks into large fragments
what occurs in a ischemia-reperfussion injury
increases ROS
Hallmarks of coagulative necrosis
"Can see cell outline but nothing else, ghostlike. occurs esp in MI and kidneys. cant see organelles, nucleus, or striations (in case of heart)"
hallmarks of caseous necrosis
"cheesy appearence, occurs in TB in the lungs and lymph nodes."
Fat necrosis
"fat destruction, pancreatic enzymes degrade fat, trauma to fatty areas"
liquefactive necrosis
"seen in bacterial and fungal infections. can occur in brain and kidney. complete digestion of the cells, results in scar tissue and cavity formation"
dystrophic vs metastatic calcification
dystrophic occurs at site of injury with normal Ca levels. metastatic occurs on healthy tissue with high Ca levles
Serous injury
"straw colored clear fluid, low in protein, EX blisters"
Fibrinous injury
"high protein content, accumulation of fibrin at injury site"
supperative injury
"abscess and pus, dead white blood cell accumulation, abscess is a walled off infection"
ulceration
eroded epithelium. gastric or epidermis
What is the definition of a free radical
Any chemical species that can independently exist with one or more unpaired electron in its outer shell
Why is O2 an ROS
it has 2 unpaired electrons
what is the most potent ROS
Hydroxyl radical
What are sources of ROS
"ETC, peroxisomes, radiation, toxins, pollution, drugs"
What are the beneficial effects of free radical
"Regulate metabolism, biomolecule turnover, microbe protection"
How is superoxide radical made
spontaneous reaction
How does the body get rid of superoxide
"superoxide dismutase converts it to H2O2, a less reactive ROS"
How does the body get rid of H2O2
1 of 2 rxns. Catalase or Glutathione peroxidase (selenium cofactor)
If H2O2 is not reduced what can happen pathologically
"Fenton Rxn- with iron to make hydroxyl radicals
When is hypochlorous acid good
Respiratory burst from phagocytes results in its production
What is the consequence of increased oxidative stress
peroxidation of cell membranes followed by increased Ca influx and cell death
How can oxidative stress be measured
"MDA, 4-HAD
Why are both E and C needed as antioxidants
"E is good for scavanging lipids, C is in the cytosol and can regenerate E."
How is NADPH related to oxidative stress
helps reduce glutathione so glutathione peroxidase can function
how is metHb dealt with
NADPH can directly or indirectly reduce it to Hb.
What in general do cytokines do
"activate cells, induce proliferation, increase cytotoxicity"
what are the 3 functions of complement
"recruit inflammatory cells, opsonization, perforation"
what are some mechanical elements of innate immuntity
"skin, peristalisis, flushing, coughing, mucus, cilia, fever"
what is a cellular element of innate immunity
"phagocytes (monocytes, neutrophils)"
What signals phagocytes in innate immunity
Pattern Recognition receptors (with aid of TLRs and Ab/complement)
What occurs during innate immunity after a PPR is activated by a PAMP
"Increased cytokines, chemokines (IL8), lysozyme, upregulation of other receptors (MHC)"
What does TLR4 recognize
LPS
What does TLR 6 bind
CpG DNA in prokaryotes
What does TLR5 bind
Flagellin
TLR3 binds
dsRNA
ICAM is a ligand for
integrins
What is the definition of a pathogen
an organism that has the potential to cause disease
what kind of immunity are B cells associated with
humoral
T cells are players in which kind of immunity
CMI and Humoral
What do T cells require to activate
a antigen presented by an APC
Why are dendritic cells special
they pick up antigens in periphery and run them to the closest node for clonal selection
what are the 2 roles of antibodies
"neutralize (as in toxins or attachment of pathogens), and target things for destruction"
What receptor is important for phagocytosis
Mannose
what ramps up phagocytes
IFN-gamma
whats a downside to innate immunity through cytokines and phagocytosis
"sloppy, lots of collateral damage"
what happens in secondary lymphoid tissue
"mature lymphocytes live there, wait for antigens and return from patrolling. antigen is also concentrated."
what does CMI require that humoral immunity does not
an APC
What is humoral immunity reactive to
free floating antigens/pathogens
What immunity targets pathogens within cells and viruses
CMI
What does CMI always require
APC and T-helper cells
What do marginal zone B cells react to
blood borne polysaccharide antigens
what do follicular B cells react to
respond to protein antigens
B1 B cells are special why
"line GI track, do not react to ingested protein but still can attack pathogens"
what is similar between Surface Ig and TCR
"both have antigen binding domains, both have variable and hypervariable regions. "
Differences between SIg and TCR
"ones on T ones on B cells, TCR have 2 chains, SIg have 4. 2 binding sites on SIg, TCR binds to MHC"
What is active immunity
"made own antibodies, via natural (infection) or artificial (vaccination) means"
What is passive immunity
"immunity came from elsewhere.
what are the cardinal features of adaptive immunity
"Specific, Diverse, Adaptable, Memory, clonal expansion, specialized, limited, discriminant"
Clonal Selection Theory States
A cell with a specific antibody exists before pathogen insult. once the antigen binds its antibody that cell then replicates and clones are produced
What factors make an antigen more immunogenic
"Larger size, protein or complexed with a protein, complex molecule"
What is an immunogen vs an antibody
immungens are antibodies that illicit an immune response
What type of receptor can bind conformational epitopes
B cells
What 3 things are required for B cells to begin secreting Ig
"stimulated by an antigen
what are the 2 kinds of light chain domains
kappa and lambda
what 2 Ig types have 4 constant domains
M and E
what holds A and M polymers together
J chains
IgA does:
"opsonization, neutralization, transporting across epithelium, can activate complement thru alt path. Found in secretions"
IgG does
"opsonization, neutralization, Sensitization for NK cells, complement, mast cell sense, crosses placenta, found in serum high concentrations"
IgM does
complement like whoa and agglutination
What gene components are in heavy chains
"V D J C, C is the tail named for Ig type"
What genes are in light chains
just V and J with a C. Kappa has more diversity than lambda
how does ELISA work
"antigens in wells, blood added, AbAb added with reactive enzyme, more IgG for antigen, the darker the well. titer is inverse of weakest dilution with a reaction"
Western blot works by
figuring out what components of a pathogen a pts antibodies are reacting too
What are the 3 functions of complement
"Opsonization, Lysis of invading organsism, Chemoattraction of inflammatory cells"
How is the Lectin pathway initiated
Mannose binding protein binds to mannose on cell surface and then acts like C1
What is the most step in all complement pathways
cleavage of C3 and the binding of C3b to the cell surface
Why is C3 so important
"opsonin, amplification. C3a is a chemoattractant"
What is classical C3 Convertase
C4b2a bound to the membrane surface
What is Classical C5 convertase
C3b2a4b
What is the attack complex
"C5b bound with C6 and C7, 8 binds once complex bind membrane and C9 creates the pores"
What non Ig molecule can iniate the classical path
C-reactive protein
How is C1 regulated
C1INH keeps C1 from over acting
What parts of compliment are anaphalaxins
C3a and C5a
what important role does C5 play
"smooth muscle contraction, degranulation of mast cells, histamine release, increased vascular permeability"
What happens if you lack end game complement components
Severe susceptibility to Neisseria
How does Alternative path get started
"C3 is hydrolized spontaneously by water binding a thioester bond. becomes iC3 which binds B, D cleaves B, you get iC3Bb, Cleaves C3 and C3b binds membrane"
What is soluable C3 Convertase
iC3Bb
what is alternative C3 convertase
C3b3bBb
How is C3 regulated in the alternative path
Properdin speeds up C3 Convertase binding
What is an antagonist for properdin
Factor H inactivates C3b by Factor I
"If someone is deficient in factor I, what happens"
iC3Bb cleaves and cleaves and cleaves until all the C3 is depleted. lots of ear infections
To what does CR1 bind
C3b or C4b
"Where can you find CR1, what does it do"
"Macrophages, neutrophils, RBC, binds tagged membrane for destruction and filtration (RBC-Spleen). enhanced function if FcR binds to Ig or when IFNg is around."
What receptor is important in the context of EBV
CR2 on Bcells and Dendritic cells
What is the function of CR2
"binds degradation products of C3, activates B cells"
CR3 and CR4 are what class of adhesion molecules
Beta-integrins
"What do CR3 and 4 bind, what do they do"
Bind iC3b (inactive C3b) lead to phagocytosis
What are characteristics of Bacterial colony morphology
"Color, size, shape, smell, hemolysis"
What are differences in membranes between eukayotic and prokayotic cells
"Bacteria dont have cholesterol, have peptidoglycan wallas"
Describe the structure of bacterial cell wall
cross linked sugars by amino acids NAM and NAG
what does lysozyme do to cell walls
cleaves B 1-4 glycosidic bonds in NAM and NAG
Teichoic and lipoteichoic acid are found where
in the gram+ cell wall.
what causes a systemic immune response like endotoxins in Gram+
Lipoteichoic Acid
What is important in the outer cell membrane of Gram- bacteria
LPS- endotoxin
"What is the ""bad part"" or LPS"
Lipid A
What is used to classify gram- bacteria
O antigen on the LPS
What is the function of bacterial Capsule
"Protects from innate IS, antigenic factors, protects against phagocytosis, adheres to surfaces"
Biofilm is
a slime layer that is less discrete than a capusle but has similar job
What is the name for the doubling time in bacteria
Generation Time
What factors affect bacterial growth
"Carbon, Nitrogen, Iron, pH, temp, osmolarity, Oxygen"
What are sterile sites in the body
"Blood, UT, Sinuses, uterus, prostate, CSF, lungs"
What conditions determine if an infection will occur
"Pathogenicity, Immune status, Route of entry, Dose of infectious agent"
What are factors determining virulence
"Structure- capsule, lps, pili.
How do eukaryotes get iron
"siderophores steal it from transferrin and lactoferrin, others have receptors for host transferrin"
How does disease occur from infection
"By products of growth- gas, acid
Exotoxins
"Proteins secreted by gram + and -.
How do endotoxins lead to septic shock
toxin is released systemically and the immune reaction is overloaded with Immune response
How can bacteria evade immune response
"degrade chemotaxins, opsonins, live inside cells, capsule can mask bound complement, phase variation"
What is the mechanism of endotoxins
"LPS binds to LPSBP, binds to CD14 or TLR, Macrophage releases cytokines"
what mediators are the big players in septic shock
"TNF-a and IL 1. cause hypotension, vascular permeability"
What are 4 types of entry of exotoxins
"AB structure, Type III, Poreforming, Superantigens"
How do AB toxins work
"A is active subunit, B is binding and mediates A's entry"
What is an example of AB toxin
"Diptheria from C. Diptheria. catalyzes ADP ribosylation of histidine on EF2, prevents protein synthesis.
What are examples of single subunit internalized exotixins
"Adenylate cyclase toxin-purtussis
Membrane Active Toxins
"Hemolysins
Membrane lytic enzymes
Clostridium perfringens- results in necrosis
Membrane active enzymes
"Staph aureus- exfoliative exotoxin- scalded skin syndrome
Superantigens
"Staph- TSST1
conjugative plasmid is
can encode for ways to transfer the plasmid to another organism
what is the lysogenic cycle
"after a bacteria is infected by a phage, the viral genome can become dormant in the chromosome and bacteria is a lyogen, with a prophage. can undergo lysogenic conversion."
Penicillin is a
"beta lactam, inhibits cell wall synthesis"
How do Beta lactams work
Bind penicillin binding proteins and prevent cross linking
How do bacteria resist the effects of a beta lactam
"lactamases cleave the lactam rings, porins mutate in gram-, cant get to wall, can change the PBP"
What is the difference in vanco
inhibits cell wall synthesis by binding D-ala
How is vanco resisted
d-ala d-ala is switched to d-ala d-lac
How does bacitracin work
prevents cell wall components from moving thru the cell membrane
what antibiotics interfere with protein synthesis
"aminoglycosides, tetracycline, macrolides"
Aminoglycosides
premature release of mRNA
Tetracycline
inhibits tRNA from binding ribosomes
Macrolides
Prevents peptide elongation
Antibiotics that are nucleic acid synthesis disrupters
"Quinolones, rifampin"
Quinolones
Block DNA gyrase
Rifampin
Inhibit DNA dependent RNA polymerase
Drugs that inhibit metabolism
"sulfonamides, trimethoprim. mess with folate metabolism"
What is the ultimate goal of intracellular signalling in IR
"activation of NFAT (tcells)
What does increased synthesis of those TFs
"Increased, CD40L, IL2, and IL2R expression"
What 2 things does ZAP do
activates PLC and RAS
What does PLC do
cleaves PIP2 into IP3 and DAG
How do immunosuppressives work on the immune system
Block calcineurin from activating NFAT
What do the ITAMs phosphorylate
P59Fyn which then P-ates ZAP
How long is the acute phase in inflammation
72 hrs
Why are some proteins negatively regulated in inflammation
"material rationing, hiding iron from bacteria"
What is CRP
"Induced APP, works with complement and increases cell survival during insult"
What does CRP bind to in invading organisms
Ca dependent binding to PCh on cell membranes of bacteria and fungus
What are the big 3 markers of acute inflamation molecularly
"ESR, CRP, SAA"
What are the components of inflammations
"redness, swelling, pain, and heat"
How does SAA work in the context of inflammation
Binds to apolipoproteins and can decrease foam cells in the system by eliminating cholesterol esters
what is alpha 2 macroglobulin
"panprotease inhibitor, lots of things can bind, cleaves them and they are phagocytized. also important in coagulation"
what is the function of haptoglobin
scavanges loose Hb from the blood
What parts of the immune system present in acute inflammation
"PMN, macrophages"
What cells are absent in acute inflammation
lymphocytes
What mediator is going to lead to a lot of vasoconstriction in inflammation
histamine
What are characteristics of transudate
"albumin, low SG, usually clear, mostly fluid"
What is exudate
"high protein content, high SG"
How do macrophages make NO
iNOS makes arginine to NO
What eicosanoids mediate vasoconstriction
TXA2 and LT
What eicosanoids mediate dilation
Prostoglandins
What are effects of IL1 and TNF in inflammation
"Decrease appetite, APP, hypotension, also increase leukocyte adherence, pro-coag, increase IL1, 8, 6"
Why is vessel constriction important in an injury site
"slows flow, more contact with inflammatory cells"
what is the order of reactions in conventiional intrinsic coagulation
"12, 11, 9, 8, 10, 5, 2, 1, 13"
what initiates extrinsic path
cell damage activates VII which cleaves X
how is ca tied to coagulation
creates a bridge between the negative cell surfaces and the negative surfaces of the platelets
How is vitamin K used in coagulation
"wakes up factors 2, 7, 9, 10, C and S."
how does warfarin (coumarin) affect vit k in coagulation
K cannot be recycled by reduction in the presence of warfarin
How is fibrinogen activated
thrombin removes the alpha and beta subunits and the fibrin monomers bind noncovalently
how does fibrin cross linking occur
Factor XIII forms covalent bonds between lysine and glutamine groups on fibrin
What effect does APC have
inactivates V and VIII
how is APC activated
endothelial cells give of thrombomodulin which activates APC
What happens in Factor V mutations
"Factor V leidin, APC cannot inactivate factor V"
How does alpha 2 macroglobulin act in coagulation
traps factors and cuts them
what are serpines in coagulation
suicide inhibitors of clotting factors
how does heparin work
changes the conformation of ATIII and enhances inhibitory effects
What does ATIII affect
"2, 9, 10, 11, 12 (intrinsic path)"
What does PTT test
intrinsic and common path of coagulation. testing for heparin effects
what does PT test
"extrinsic path, Factor VII and the effects of coumadin."
What bacteria are catalase positive
staph
How can you tell the difference between strep and staph
strep are catalase negative and staph is catalase positive
pt has a catalase + and coagulase + bacteria what is it
staph aureus
what strains of staph are coagulase -
staph epidermis and staph saprophyticus
Identifying characteristics of staph aureus
"gold colonies, beta hemolytic, coagulase +, protein A (binds Fc) capsule, grows in high salt"
What is a key pathogenic feature of staph aureus
can cause osteomyelitis
staph aureus toxins present how
"exfoliatin toxin- scalded skin syndrome
why arent PCNs effective against staph
have beta lactams in 90% of strains
what is typically used to fight staph
"pcn derivatives, vanco against mrsa"
What immune factors combat staph infections
opsonizing IgG
what does staph epidermidis look like
"white non hemolytic colonies, coagulase negative. high mw slime"
what is the key factor for identifying epidermidis vs saprophyticus
sapro is resistant to novobiocin
what are the medically important strains of strep
"A, B and enterococcus"
what does alpha hemolysis mean
incomplete hemolysis
what is beta hemolysis
complete hemolysis
Key factors in GAS
"(strep pyogenes), very sensitive to bacitracin, M protein and lipoteichoic acid in pilli. epitopes are similar to cardiac muscle"
what does streptolysin S do
"lysis leukocytes, platelets, and RBC"
Strepolysin O does
all what S does plus its immunogenic
How do you diagnose a GAS
"Gram +, catalase -, sensitive to bacitracin"
How can u treat GAS
PCN works
Strep agalactiae is different how
resistant to bacitracin
why is strep agalactiae important
#1 cause of neonatal sepsis and meningitis
Dx of enterococcus
"Gram+ in pairs, non hemolytic, grow in high salt, high temp, and survive bile"
how do you treat entero
"usually need more than one, usually resistant to vanco"
Strep pneumo
alpha hemolytic
Enterobact
"are everywhere, cause sepsis, UTI, GI, pulm. all ferment glucose, oxidase negative"
E. Coli
UTI king. ones that cause GI probs have a extra plasmid.
Shigella
bloody diarrhea
salmonella
gastroenteritis
klebsiella
necrotizing pneumo
Mac plates
"inhibit gram+, lacotose fermenters will be pink, non lactose will be colorless"
EMB plates
"inhibit gram +, ecoli are green, others are pink"
How does Ecoli bind to the gut
pilli
what organism has a lot of capsule
klebsiella
what stimulates early proliferation of CLPs
IL7
what happens if you lack IL7
SCID- Bubble boy
How does heavy chain recombination occur in B cells
"occurs between proB and preB stages. VLD chains rearrange, DJ first VDJ second. "
When does light chain rearrangment happen and how
"in preB stage after successful heavy chain mix. and mu+heavy chain is expressed w a surrogate light chain. small preB starts, K than L light chain rearrangement"
How does negative selection of B cells occur
"in bone marrow if it reacts to self antigen can be killed, anergy, or will edit its receptor"
once B cells migrate out what happens
can start expressing IgD along with IgM
How do T cells mature
"Begin as double negative (no CD4 or 8) beta chain made first in TCR, with rearrangment, after alpha is rearranged and added. Become double positive, by random occurance meet MHC 1 or 2 and then express 4 or 8 depending on what they hit"
if thymocytes fail to recognize MHC what happens
signaled for death (fail to positive select)
CD1
thymocytes and langerhans cells
CD3
associated with TCR
CD4
"Thelper cells, hook MH2"
CD5
costimulation on T cells
CD8
CTL recognize MHC1
CD10
preB cells
CD19
all B cept Plasma
CD20
all B cept plasma
CD21
EBV receptor on B and dendritic
CD23
B cells
CD79
all B cells
What is the intial contact of rolling T cells
L-selecting binds CD34 on HEV (or glycam)
What is the second contact in rolling T cells
LFA1 onto ICAM
what is the costimulatory signal
CD28:B7.1 (CD28 on TCell)
what happens if Tcell doesnt get costim signal
anergy
what is CD40L
"binds CD40 on macrophages and B cells, causes them to PAD and get ramped up. also stims class switching to IgG"
What does IL2 do
"made by t cells, autocrine signal. will PAD"
How do Th1 cells form
IFN-gamma/IL12 signals
What do TH1 cells do
make IFN gamma. important for chronic inflammation. make TNF
What do TH2 cells do
"IL4 induces, make IL4,5, and 13. important for allergy and parasites. produce more IL10 than TH1"
what happens when a CD4 cell meets a Bcell with antigen on an MHC
CD40 and CD40L interaction. CD4 cell secretes cytokines that make B cell PAD and secrete/class switch
what makes an antigen T cell independent
"all sugar, no protein, can stimulate Bcells on their own, no memory tho"
what effects do TH! cells have on macrophages
"Increase NO, ROS, Proteases, MHCII, B7, CD40, TNFR.
What do macrophages release upon signalling from a TH1 cell
more IL 12 which makes more TH1 cells.
what is needed to activate NK cells
IL2
What secrete interleukins?
"Leukocytes, also they stimulate leukocytes"
What cytokines regulate innate immunity
"Type I IFN, TNF-a, IL 1, IL 6, IL, 12, IL 10"
What are the 2 groups of Type I IFN
A and B
What secrete INFa
Mononuclear Phagocytes
What secrete IFNb
Lots of cells
What are Type I IFNs important for
Viral infections
What transduction path to Type I IFNs follow
JAK STAT
what is an ISRE and what stimulates it
"interferon sequence response element, IFN"
What is the response to Type I IFN
"upregulation of: oligoadenylate synthase, dsRNA serine/threonine Kinase, RNAses, Increase MHC I, upregulate IL 12r (increases Th1 proliferation, enhance NK cell cytotoxicity"
What is the main goal of an IFN response
inhibit proliferation of viruses and enhance their destruction
How can IFN be used therpeutically
"Cancer, Virus treatment"
What are the toxic effects of IFN
"Flu like, fever, chills, malaise, myalgia, headache, depression"
What are Pro inflammatory cytokines
"TNF, IL1, IL6, chemokines"
What is the primary response to gram - bacteria
TNFa
What releases TNFa
"activated mononuclear phagocytes (can also be released by Th1, NK, and Mast cells)"
What augments production of TNFa
IFNg
What does TNFa do
"upregulates selectins, VCAM and ICAM, increases chemokine secretion by macros, promotes clot formation, induces fever (thru prostoglandins), cachexia, and increases APP"
Prolonged TNFa secretion results in
sepsis
What are the hallmark symptoms of sepsis
"hypotension, shock, disseminated intravascular coagulation, crazy blood sugar levels"
What do CC affect
"monocytes, lymphocytes, eosinophils"
What do CXC affect
neutrophils
What is the function of chemokines
"Recruit cells, promote angiogenesis"
what produces IL 12
"mononuclear phagocytes, macrophages, dendritic cells,"
what does IL 12 do
"activates NK, lymphocytes, stims IFNg production, Th0->Th1, increases cytotoxicity"
what does IL 10 do
opposes everything IL 12 stands for. inhibits macros and dendritics
what makes IL 10
activated macros
What helps shut down T cell activity
IL 10
What is the principle purpose of IL 2
Moving T cells from G1 to S and maintaining T-regs
What makes IL 2
"CD4 (Th0, Th1) and lesser so CD8"
What stimulates IL 2 release
stimulation by an antigen
What antiapoptotic factor does IL 2 affect
increases Bcl2 in T cells
What other factors does IL 2 increase
IFNg and IL 4
What IL is redundent with IL2
IL 15
What occurs in IL 2 toxicity
Hypotension and edema
what does IFNg do
"Stims B cell class switching (IgG2), inhibs Th2 growth, activates macros, Increases MHC, activates NK cells"
G-CSF
"used in myelosuppressive chemo, fights granulocytopenia and regens neutrophil populations"
What are the side effects of G-CSF
"splenic rupture, allergic reactions"
GM CSF
"used after bone marrow transplant, helps repopulate neutrophils and monocytes"
What are the side effects of GM CSF
"bone pain, fever, edema"
Peptostreptococcus
"Gram + cocci, normal flora in mouth and colon. can be pathogenic in lots of places."
Clostridium
"Gram + bacilli, forms spores"
Bifidobacterium
Gram + non spore forming.
What can cause brain absesses
"peptostreptococcus, fusobacterium, prevotella"
Actinomyces
"Gram + bacilli, long filiments, can be mistaken as fungus. aerotolerant. found in mouth and skin. worry about invasion after oral surgery"
Baceroides
"Gram - rods, found in colon, worry about endocarditis and abd infections"
B. Fragilis
can cause endocarditis
Prevotella melaninogenica
"capsule, gram- rod, found in mouth, causes inf above diaphragm"
Prevotella bivia/disiens
"gram- rods, GYN infections"
Porphyromonas
Gingivitis and dental abscess
Fusobacterium
"gram - rods, pointed ends, mouth, colon, vagina. in mixed infections"
Lactobacillis
"Gram- rod, falc or obligate anaerobe. non pathogenic"
mobiluncus
"Gram+ but shows up Gram-, bacterial vaginosis"
Veillonella
"Gram - cocci, in mouth, associated with bite wound infections"
C. Perfringens
"Gram+ rod, spores, double hemolysis, cellulitis, myonecrosis, food poisoning"
what toxin do all strains of C. perf produce
alpha toxin
what does C. perf Beta toxin do
"necrotizing, produced by Type C strain"
What do C. perf Group A do
"food poisoning, produce enterotoxin, causing diarrhea"
What does tetanus toxin do
"Inhibits GABA, produces spastic paralysis"
Risus Sardonicus
Tetanus smile/grimace
Tetani back spasms
opisthotonus
C. botulinum produces
flaccid paralysis by inhibiting acetylcholine release
What is C diff toxin A
"diarrhea, and chemotaxis of neutrophils by increases cytokines"
What is C diff toxin B
damages cells by disrupting actin
How does chlamydia stay alive in the cell
controls vacuole movement to avoid lysosomal fusion
What is super creepy about listeria
once it invades the cell it disrupts actin and then uses the actin to move cell to cell
What is Type 3 secretion?
Make a pore in the membrane and then can control gene expression. shigella does this
The body requires ______ to clear intracellular bacteria infections
exogenous therapy (antibiotics)
What does chlamydia pneumo bind to avoid destruction
"binds complement receptor rather than IgR. Cr's don't produce Oxidative burst, IgR does"
What cell type do Rickettsia orgs usually invade
endothelial cells
Rickettsia prowazekii
"louse borne typhus, vasculitis, usually appears in disaster areas"
Rickettsia tsutsugamushi
"Scrub typhus, mite transmitted"
Coxiella burnetti
"CV disease, Q-fever, livestock contamination, resists lyso enzymes. can lead to endocarditis"
Ehrulichia chaffeenis
"monocytic ehrlichiosis, lone star tick, leucopenia"
Chlamydia trachomatis
"GU tract, prevent lysosomal fusion, move vacuole to golgi or ER"
Chlamydia Pneumo
"community acquired pneumo, athlersclerosis, might have alzheimers connection"
Chlamydia psittocosis
"causes psittacosis, sever pneumo"
mold
filamentous form
yeast
single celled form
dematiaceous fungi
"black/brown fungus, opportunistic, phaeohyphomycosis"
Hyphae
filaments/tubular structures in molds
coenocytic
nonseptated
pseudohyphae
formed by incomplete budding of yeast cells
mycelium
mass of intertwined hyphae
vegetative mycelium
absorbs nutrients
aerial mycelium
reproductive
Conidia
asexual spores
conidiophores
support chains of conidia (aspergillus)
sporangiospores
sac like structure (in zygomycetes)
Blastoconidia
budding yeast cell
Arthroconidia
conidia formed by fragmentation of hyphae
Cutaneous mycoses
dermatophytes (ringworm)
Superficial Mycoses
Tinea versicolor
Systemic mycoses
"internal organs, opportunistic"
Endemic mycoses
dimorphic
How is candida variable
"its yeast when its normal flora, when it becomes an infection it becomes a mold"
What happens in a systemic mycosal infection
"inhale/ingest a mold, then becomes a yeast in the system"
Echinocandins
"cell wall synthesis. inhibit B 1,3 glucan bonds. good against candida and aspergillus"
Types of ergosterol synthesis inhibitors
"allylamines, azoles, polyenes"
Allylamine
"inhibit squalene epoxidase, build up squalene and cell dies. step in ergosterol syn. accumulate in nails. terbinafine"
Polyenes
"amphotericin B, bind to ergosterol and form pores"
Azoles
P450 related (liver tox) ketoconazole (dimorphics) and non-ketoconazoles (cutaneous)
triazoles
greater specificity for fungal enzymes. less effects. fluconazole. broad spectrum.
Amphotericin B has what side effects
"liver toxicity, generates free radicals, renal impairment"
Nystatin
alternative to amphotericin B
Flucytosine
needs to enter cells and then disrupts DNA synthesis. used against cryptococcus. usually coupled with another drug
Griseofulvin
binds keratin and prevents infection
What are the top 3 killers
"HIV, TB, Malaria"
What is the life cycle of Borellia burgdorferi?
"2 years, larva, nymph, adult. nymph stage is the infective stage"
What is the lyme disease rash called
Erythemia Migrans
Symptoms of Lyme disease
"Heart Block, acrodermatitis, lymphocytoma, morphea"
What does Ehrlichia chaffeenis infect
monocytes
What does Anaplasma phagocytophilium infect
granulocytes
What disease shows a maltese cross on blood smear
babesiosis
What is a key factor in classifying a zoonotic infection
transmitted among vertebrates only
what is a key radiologic factor in the dx of anthrax
mediastinal widening
What are the steps in wound repair
"Injury, thrombus formation, acute inflammation, cellular invasion, synthesis of repair components"
What are the 4 steps of the hemolytic stage of healing
"Platelet aggregation, cytokine secretion, clot formation, "
Cytokines released by platelets
"PDGF, EGF, TGFb, IL1, LTB4"
what is FDB
fibrin degradation products. chemotaxin for neutrophils
what peptide do neutrophils bind
n formyl methionine on microbes
what form of PDGF is important in athlerosclerosis
BB
TGFb
"can be stim, inhib, or both depending on siyuation"
TGFa
repithelialization
EGF
reepithelialization
FGF
prolif of fibroblasts
VEGF
"angiogenesis, autocrine signalling"
KAF
"keratinocyte growth factor, reepitheliazation"
what do laminin and vitronectin do
"cell migration, form endothelium, angiogenesis"
"What cells are required to provide strength, regulation and structure during angiogenesis"
Pericytes and smooth muscle cells
What causes angiogenesis
"Injury, tumor secreting growth factors"
Vasculogenesis
"BV assembly during embryonic development, make from ECP/angioblasts"
Angiogenesis
BV formation in adults. form from already existed vessels or recruited EPC from bone marrow.
What stimulate EPC
"chronic inflammation, fibrosis, tumor growth, ischemic tissue"
Hemangioblasts
link hematopoietic and vascular systems during development
Angioblasts
"Proliferate and migrate to periphery. differentiate into endothelium, pericytes, lymphatics, SMC"
What steps are required for angiogenesis
"Vasodilation by NO, increased vasc permeability by VEGF, Degrade ECM, Migrate EPCs"
Where are VEGF receptors
"on epithelium, VEGFR2"
Ang 1 recruits
periendothelial cells (Tie2)
Ang1/Tie2
Maturation and maintains quiesence once maturation occurs
Ang2Tie2
"loosens preexisting endothelium, making them more responsive to other signals"
What is unique about tumor vessels
very irregular and leaky and grow continuously
Angiogenic Switch
"When tumors switch to angiogenic phenotype, when it is turned on prognoses is poor"
What does a malignant cell need to grow
"Growth signals, insensitivity to inhibition, invasion and metastasis, limitless growth potential, evasion of apoptosis"
VGEFR1
physiologic angiogenesis receptor
VGEFR2
receptor in tumor angiogenesis
VGEFR3
important in lymphatics
Carcinomas spread via
lymphatics
Sarcomas spread via
hematogenous routes
What are the 3 main causes of chronic inflammation
"persistant infection, persistant toxins, autoimmune disease"
What are the main cells of chronic inflammation
macrophages and lymphocytes
What is the hallmark of chronic infection
tissue destruction
how does the maturation of macrophages occur
monoblasts are released from marrow into the blood (monocytes) then into the tissues (macrophages)
what are the 3 ways macrophages accumulate in an infection site
"recruitment, proliferation, immobilization"
what do glucocorticoids do
"stop transcription of IL6 and IL1, and suppress COX and PLP"
"Pentoxifulline, thalidomide"
suppress release of TNFa
Infliximab
"blocks TNFa, can lead to lymphomas though."
3 possible outcomes of infection
"Resolution, Regeneration, Repair"
Resolution
"Original architecture stays intact, infection is cleared with no morphology changes"
Regeneration
"Cells proliferate to replace lost tissue, no changes in structure compared to original tissue"
What cell types cannot regenertate
"Permanent Cells, such as mycocardium and CNS cells. Scarring occurs and lose function in that area"
Repair
Body brings in materials to fix an area.
What creates a scar
loss of cellularity in an area and is filled in by collagen
What is Fibrosis
large amounts of collagen being laid down in an area of damage. TGFb important in the formation of fibrotic areas
Matrix metalloproteinases
zinc dependent enzymes that allow degradation events
in the healing of wounds by 2nd intention what occurs
"massive amounts of granulation tissue, lots of collagen deposition that can result in contracture"
What local factors influence healing
"persistent infection, blood supply, movement, irradiation, drugs (NSAIDS, Steroids)"
What systemic factors influence healing
"age, nutrition, metabolic disease, malignancies, systemic drugs."
LOX makes
leukotrienes
COX makes
thromboxanes and prostaglandins
What chemicals can produce erythema
PGE2 and PGI2
What do prostaglandins do
"produce fever, increase renal blood flow, GFR, mucus secretion in GI, inhibit stomach acid and thrombosis."
TXA2
induces platelet aggregation
at low doses what do NSAIDs inhibit
selectively inhibit TXA2 (stop platelet aggregation)
What happens as the NSAID dose increases
begins to inhibit PGI2 (therefor promotes clotting)
What are the 4 As of NSAIDs
"Analgesic, Anti-inflammatory, Antipyretic, Antiplatelet."
What are general contraindications of all NSAIDs
"ASA hypersensitivity, Liver dysfunction, Renal dysfunction, Alcoholism, Pregnancy/Breastfeeding"
Salicylates
"have all 4 As, increase resp, change acid/base, irreversible cox, high protein binding"
What are the contraindications of salicylates
"gout, bleeding disorders, chicken pox, flu"
what occurs in salicylate tox
"tinnitus, vertigo, confusion, delerium, GI bleed, N/V, heptatotoxicity"
What is different about diflunisil
"no antipyretic, used for severe, inflammation, much more anti-inflam capacity, can't be used with asthmatic pts"
Para-aminopheno derivatives
"tylenol, low anti-inflammatory capacity. lower protein bound, can be used where ASA can't."
Entercoccus spp
Amp +/- gent
S. Aureus
Nafcillin
MRSA
Vanco +/- gent
S pneumo
PCN
PCN resistant S pneumo
Vanco+ceftriaxone
N. gonorrhoeae
PCN
N. meningitidis
Cefuroxime
C. diff
Metronidazole
Chlamydia
Macros
T pallidum (syphillis)
PCN
Bacteriodes
Metronidazole
E coli
3rd gen cephs
Propionic Acid Dervis
"Ibuprofen, naproxen"
Ibuprofen
"4 A's, reversible platelets, Comp Inhib Cox, No interactions. ADR: headache, ocular issues"
Naproxen
"Inhibs PMN migration, has ototoxicity, 4 As, reversible platelets"
Heteroaryl Acetic Acid
"Ketorolac, Tolmetin, Diclofenac"
Ketorolac
"4 As, reversible, Potent COX inhib, drowsiness"
Tolmetin
"4 As, little occult blood loss, RA, OA, No competition, Sever GI side effects and tinnitis"
Diclofenac
"3 As, no platelets, RA, OA, Spond, reduces Arach acid, High FP effect, sin rash, some competition"
Enolic Acid
"Piroxicam, Phenylbutazone"
Piroxicam
"4 As, RA, gout, OA. Inhibs neutros, some competition, GI ADRs"
Phenylbutazone
"very toxic, RA, acute gout, lots of competition, Nephritis, hepatitis, anemia"
Indomethacin
"3 As, RA, Spond, acute gout, close ductus arteriosis, most potent COX inhib, Significant bleeding, depression, psychosis"
Sulindac
"Prodrug for indomethacin, easier on pt"
Celecoxib
"selective Cox2 inhib. 3 As no platelet, P450 mets, very little ulceration"
Type 1 Hypersensitivities
"Rapid, IgE, no IL12 and no IFNg, no macros"
What Il do Th2 cells produce
"4,5,6,10,13"
What does IL4 do
Induces class switching to IgE
What Il activates eosinophils
IL5
What is the first mediator of a Type 1 response
"Histamine, H1 is for allergies"
What occurs during the second wave
"chemokines, cytokines, PGD2, LC4/D4, occur leading to neutrophil invastion, brochospasm, constriction and innflammation"
Uticaria
Hives
What is the infective state of protazoa
Cyst form
What is the pathogenic state of protazoa
trophozite
What are the 2 types of playhelminths
Cestodes (tape) and trematodes (flukes)
What is a common sign of parasite infection
Charcot-Leyden Crystals
What are the major HLA groups looked at in matchin
"A, B, DR. Maternal and paternal total of 6 matches"
How do we know graft rejection is immunologic
animals sensitized to donor MHC react to a transplant as they would a repeat pathogen
Hyperacute Rejection
"AB mediated. Host must be sensitized, very very quick. Type II Hypersensitivity with vasculitis."
Acute Rejection
"Necrosis, hemorrhage, endothelitis, CD4 and 8 and macros. Necrotizing vasculitis. Swollen endothelium and parenchyma"
Chronic Rejection
"Thickening of vasculature, Smooth muscle prolif, narrowing of intima by fibrosis. no necrosis, has atrophy."
Acute GVH
"Necrosis, Jaundice, diarrhea, rash"
Chronic GVH
"Fibrosis, Atrophy, Jaundice, Eso Strictures"
What is the immune response in GVH
Donor T cells release cytokines which activate host macros and NK cells to do damage to the host.
How does cyclosporine work
blocks IL2 transcription by binding cyclophilin and then it blocks calineurin and NFAT cant dephosphorylate
How does Tacrolimus work
"Binds FK binding protein, binds calcineurin like cyclo"
Sirolomus
"Binds immunophilins and inhibs calcineurin, blocks B cell prolif"
Mycophenolate mofetil
"stops synthesis of purines, stops b a t mitosis."
Corticosteroids
"Interferes with cell cycle of lymphoid system, inhibs inflammation"
DNA Viruses
"Host polymerase, very durable, replicate in nucleus, genome is template for mRNA"
+ strand RNA
"genome is mRNA, encodes RNA dependent RNA polymerase. Transcription makes template for mRNA and genome. Makes a poly protien"
Infectious Genomes:
DNA and + strand RNA viruses have genomes that can initiate an infection alone
- Strand RNA
"RNA dependent RNA poly is a core protein, genome is template for mRNA, transcription makes template for genome"
Retroviruses
"+ strand RNA is transcriped into DNA. RNA dependent DNA pol is a core enzyme, transcription of cDNA makes genome and mRNA"
PCN
URI
Nafcillin
PCN resistant Staph
Amoxicillin
URI
Ticarcillin
Pseudomonas
Piperacillin
4th gen PCN
Imipenem
Beta-lactam
Vancomycin
Gram +
Aztreonam
Gram -
Cefazolin
1st gen Ceph
Cefoxitin
2nd Gen Ceph
Cefacor
2nd Gen Ceph
Ceftriaxone
3rd gen Ceph
Cefotaxime
3rd gen Ceph
Nalidixic Acid
Quinolone
Ciprofloxacin
Fluoroquinolone
Ofloxacin
Fluoroquinolone
Gatifloxacin
3rd gen FQ
Levofloxacin
3rd gen FQ
Sulfamethoxazole
Sulfonamide
Sulfisoxazole
Sulfonamide
Sulfasalazine
Sulfonamide
Sulfacetamide
Sulfonamide
Doxycycline
Tetracycline
Minocycline
Tetracycline
Gentimycin
AMG
Tobramycin
AMG
Amikacin
AMG
Streptomycin
AMG
Spectinomycin
AMG-related
Erythromycin
Macrolide
Azithromycin
Macrolide
Clarithromycin
Macrolide
Clindamycin
Misc
Metronidazole
Misc
Quinupristine
Misc
Linezolid
Misc
Chloramphenicol
Misc
What in general are AMGs for
Serious Gram - infections
What drugs inhibit the 50s subunit in bacteria
"Macrolides, chloramphenicol, linezolid"
What antibacterials are contraindicated for pregnancy
"AMG, tetracyclines, FQ, Sulfonamides"
What are the DOC for Pseudomonas
"3 and 4th gen PCN, Ticarcillin, pipercillin"
What lactamase inhib is paired with pipercillin
tazobactam
What is paired with clavulanic acid
amoxicillin
What can you use for a gram - infection with a PCN allergy
Aztreonam
What is the DOC for drug resistant gonorrhea
Ceftriaxone (3rd gen)
what are the 2nd gen FQs
"Cipro, ofloxacin, lomefloxacin"
What is an important consideration in prescribing Quinolones
can lead to cation toxicity if taking mineral supplements and competes for the P450 system with other drugs
In general 2nd gen FQs are used for
"Gram -, STD, and neutropenic pts"
What is the difference between the 2nd and 3rd gen FQs
Better Gram+ coverage in 3rd gen
Levofloxacin is a
3rd gen FQ
What consideration needs to be taken with 1st generation quinolones
want to acidify urine to promote secretion
What typically are sulfanamides restricted to
opthalmology and uncomplicated UTI E.coli (complexed with trimethoprim)
What is the MOA of sulfanamides
block folic acid synthesis
What drugs can have the ADR of kernicterus
Sulfonamides
Pneumocystis jiroveci
Sulfamethoxazole and Trimethoprim
Prostatitis in young males
oflaxacin
Prostatitis in older males
trimethoprim/sulfonamide
What is the MOA of tetracyclins
Bind the 30s subunit. block tRNA from binding the A site
What is the spectrum of tetracyclins
"pretty broad, DOC for lyme disease, rickettsiae."
What is the MOA of AMGs
binds to the AUG site of the 30s stops all protein synthesis.
What is the worst part about AMG
Ototoxicity
What is the primary spectrum for AMG
gram -. complexed usually with PCN
What is used to treat TB
Streptomycin
Typically what is used to treat pcn resistant chlamydia
Eryrthromycin
What is the DOC for C diff
Metronidazole
What drugs are good against Vanco resistant gram+
"Quinupristine, Linezolid"
What strain of HIV predominates in the US
HIV-1 Group M Clade B
What is Group N HIV
Recent combination of SIV and HIV-1
Into what family and genus of virus does HIV fall
"Retrovirus, Lentivirus"
What is unique about HIV 2
Less virulent and infected pts show resistance to HIV1. slower progression to AIDS. Has 5 clades
In the early stages of infection what can describe HIV
macrophage trophic virus R5
When does HIV start producing syncitia
later stages when it is T cell trophic
When is HIV more cytopathic
Late stage
What is the receptor that makes HIV trophic for T cells in the late stage
CXCR4 receptor (X4 virus)
How many copies of the genome exist in a virus
2
What surrounds the genome
nucleocapsid protein p7 also contains the RT
What is the capsid protein
p24 surounds the nucleocapsid
What does the matrix protein do
p17 is involved in the assembly of the virus
what are the 2 parts of the envelope
"gp120, the globular head and outermost part
what does gp120 do
is responsible for attachment
what does gp41 do
mediates fusion of the virus and host cell
what does env encode for
gp120 adn gp41
what does pol encode
"enzymes for replication
what does gag encode
"Structural proteins, matrix, capid, nucleocapsid"
what genes overlap
"gag and pol, gag makes own protein, gag=pol polyprotein"
how is env transcribed
"makes gp160... cleaved then into gp120, gp41"
what does tat encode
transactivates transcription (turns on ts)
what does rev encode
promotes mRNA export to cytoplasm
nef
"contributes to virulence. if missing nef, virus is attenuated, needed to progress to AIDS"
vif
"helps assembly, blocks antiviral proteins"
vpu
"inhibits tetherin, allows virus to be released from cell"
vpr
transports cDNA to nucleus
what does the LTR do
"promoter, enhancer sequences for host TS factors"
how does HIV attach to a cell
"gp120 binds CD4 and allows CCR5 to bind gp120 via V3, which allows gp41 to interact with plasma membrane and allow fusion"
what can prevent infectivity of HIV
if the virus doesnt cleave the polyprotein
What creates quasispecies within an individual
Lots of mutations
What can the mutations in HIV do
"Escape IR, resist ARV, change R5-X4, allow syncytia formation"
What receptors do T cells express that are attachment site for HIV
"R5, X4"
NRTI
nucleoside RT inhibitors
What must occur to activate an NRTI
must be phosphorylated by a cellular kinase
NRTI MOA
"binds to RT and inhibits RT, results in chain termination. Zidovudine"
NNRTI (non nucleoside RT inhibitor
No phosphorylation needed. binds to RT at non-active site and inhibits. Nevirapine
Protease inhibitors
"Binds HIV protease, virus cant mature. Saquinavir."
Fusion inhibitors
"enfuvirtide, binds gp41 and virus cant fuse"
CCR5 inhibitor
"Maraviroc, binds CCR5. alters conformation, can drive virus to become X4"
Integrase inhibitors
"Raltegravir, does what it says"
What happens in the primary infection
"high viral loads, flu/mono like symptoms. infecting CD4 cells and CD8 cells kill off productive CD4"
During reinfection what areas are hit the hardest
MALT and GALT. areas with a lot of T cells
How are T cells depleted
"Directly via the virus, apoptosis induced by the virus, or from CTL cells"
What is being screened for in an acute infection
"antibodies to p24, viral load (PCR), p24"
what happens if HIV is opsonized by AB
"Nothing, is phagocytized but survives in the cell"
What is ARC
"AIDS related complex- CD4 is 500-200. lymohadenopathy, fever, weight loss, malaise. some OI"
What are AIDS defining illnesses
"Thrush, Karposi, Dementia"
What is the biggest obstacle in developing an HIV vaccine
"target would be gp120, but is very variable"
How would using humoral immunity help defend against HIV infection
T cells would be activated earlier and be able to make the set point of the virus very low and manageable
What are indicators for testing for HIV
"other STDs, Opportunistic infections, odd lab findings"
What does the CDC recommend for testing
"Routine testing for 13-64 yo in health care settings, opt out rather than opt in, no seperate consent for test, no counseling, repeat testing at discression of dr"
What are the big 4 symptoms of acute HIV
"Fever, Lymphadenopathy, Pharygitis, Rash"
What other things are in the differential for HIV
"Group A step, Mono, Malignancy"
What are indicators for beginning ART
"Hx of AIDS defining OI, CD4 350-500, pregnant women, HIV associated nephropathy, Hep B"
What is the main goal of ART
restoration and or preservation of immunologic function
what is the typical regimen for ART
NNTRI/PI and 2 NRTIs
what does it mean to be a preferred therapy
"Optimal efficacy and durability, favorable tolerability and toxicity"
What is the preferred therapy
efavirenz/tenofovir/FTC (atripla)
what is the preferred therapy for pregnancy
LPV/ZDV/3TC
what are non infectious body fluids
"urine, feces, vomitus, saliva, sweat, tears"
what are infectious body fluids
"blood, vaginal secretions, semen, breast milk"
What happens in lymph nodes during HIV infection
"Germinal centers are hyalinized, ,follicles involute, B cell activation leads to hypergammaglobulinemia"
What is PGL and how does it present
"persistent generalized lymphadenopathy. 2 non inguinal nodes, non tender, follicular hypoplasia"
What kind of lymphoma is common in AIDS
"Non-hodgkin's, 80% systemic, 20% CNS"
What occurs in stage 4 HIV
"OI, pneumocystis infections, neoplasms, HHV8, bedridden"
What diseases do you become susceptible to at CD4 500
"KS, Lymphoma, TB"
What diseases do you become susceptible to at CD4 250
"Pneumocystis, toxo, Candida"
What diseases do you become susceptible to at CD4 100
"CMV, Crypto, mycobacterium, encephalopathy"
PML
progressive multifoccal leukoencepphalopathy. autoimmune demyelinating disorder
What problems arise in the kidneys in HIV
Focal segmental glomerulonephritis
Oncogenes
Growth promoting
Tumor Suppressor
Growth stopping
what kind of mutation typically occurs to create tumors
over expression of growth factors due to gene amplification
C-erb-B1
80% of lung cancer
What happens when Ras is mutated
cannot cleave GTP and signal transduction cannot be stopped
What GAP do
enhances Ras GTPase activity
What happens if Myc is mutated
amplifies replication signal
What happens in a Cyclin D or CDK4 mutation
when they bind together in G0-G1 phase they are permanently activated and cannot stop cell cycle by continuous inactivation of Rb
Oncogenes: Dominant/Recessive?
Dominant
Tumor Suppressor Genes: Dominant/Recessive?
Recessive
What does p53 do
"Monitors cell and DNA, can inhibit replication and helicase, inhibits Rb, triggers apoptosis, induces p21"
What does TGF-b do
suppress c-myc
Desmoplasia
"abnormal collagen deposition in a neoplasm, makes tumor rigid"
what are the characteristics of a benign neoplasm
"Focal, confined, uniform, matured cells, diploid"
what are characteristics of a malignant neoplasm
"rapid growth, immature cells, anueploid, can metastasize"
Hamartoma
proliferation of cells native to an organ type
Choristoma
"Developmental, proliferation of cells not native to the organ. (pancreatic tissue growing in stomach)"
at what size does a tumor become palpable
"1 cm, about 30 doublings"
why are more than one form of chemotherapy used
there is always a chance that there is at least 1 resistant cell that will be selected for if only 1 therapy is used.
What is Primary/Induction Therapy
"Used mostly for liquid tumors, used when disease is systemic"
Adjuvant therapy
When disease has spread outside of the locally controlled area. Used after surgery
what do you have to be concerned with combination therapy
"use different MOA, dont combine toxicities, different metobolism"
ADR: Antracyclines
Cardiac tox
ADR: Taxanes
Neuro tox
ADR: Alkylating
"Cardiac tox, 2dary malignancy"
ADR: cisplatin
renal and neuro tox
ADR: cyclophosphamide
Hemorrahagic Cystitis
Cell type+OMA=
benign (exception- melanoma)
Cell of origin for Sarcoma
mesenchymal cell
Cell of origin for carcinoma
epithelial cell
what is differentiation
Extent to which tumor resembles the tissue of origin
What level of differentiation comes with a poor prognosis
the less differentiated the worse the prognosis
Anaplasia
"lack of differentiation, usually more aggressive"
what are you going to see microscopically on a malignant neoplasm
"crowded glands, high N/C, irregular shapes, generally ugly looking"
How long is a course of radiation therapy
everyday for 1-9 weeks
IMRT
intensity modulator radiation therapy. MCL changes throughout tx to modulate radiation.
IGRT
image guided. can visualize tumor as you treat.
What is a brag peak
term in proton therapy. proton deposits most of its energy in the treatment site and then leaves the tissue. no residual radiation
what does vWF bind
GP Ib/IX/IV
What is contained in platelet granules
"ADP, serotonin, platelet factor 4"
What allows the platelets to bind together
GP IIb IIIa receptor connects with fibrinogen after a conformation change
Thrombocytopenia is
low platelets
What are the general causes of thrombocytopenia
"too much destruction, too little proliferation, sequestering in the spleen. hemodilution/spurious"
what is the 2nd step after a blood smear
"bone marrow, look at megakaryocytes. none- cancer/aplasia. normal- platelet destruction"
If everything looks normal what is next in dDx
"DIC, TTP, SLE, ITP (very last thing)"
What are causes of decreased platelet production
"leukemia, cancer, fibrosis, infection, TB, drugs, chemicals"
Congenital Thrombocytopenia
"absent radius syndrome. low platelets, weird skeletal things"
May Hegglin Anomaly
"Auto Dominant, purpura, giant platelets, Dhole bodys in neutros"
Wiskott Aldrich
"IgM deficiency, allergies/eczema"
what can cause increased platelet destruction
"autoimmune, lymphoma, drugs, infections, DIC, TTP, HUS"
ITP
IgG autoantibodies against GP IIb/IIIa
TTP
"vWF multimers are supposed to be cleaved by ADAMTS13, pt has huge vWF clumps and start forming thrombi"
How does TTP present
"Thrombocytopenia, Microangiopathic hemolytic anemia, headaches, renal dysfuntion, fever, schistocytes"
HUS
"hemolytic uremic syndrome. looks like TTP, caused by gastroenteritis"
NAIT
"neonatal alloimmune thrombocytopenia. mom is PLA neg and baby is positive, moms antibodies cross placenta"
Type II HIT
"heparin induced, antibodies bind to PF4-Heparin complex and cause thrombosis"
What 4 factors are used in platelet aggregation testing
"epi, collagen, ADP, Ristocentin"
Type I vWF disease
decrease in all polymeric forms of vWF
Bernard Soulier Syndrome
"Auto recess. Def of GP Ib IX. platelets cant stick to walls. Large platelets, severe bleeding, rsitocetin decreased"
Glazmann Thromboasthenia
"weak platelets, auto rec, GP IIb IIIa disorder, platelets dont adhere to eachother. ristocetin is normal, others are decreased."
Storage pool disease
can either lack granules or cant release them
Petechiae
platelet/vessel disorder
mucosal bleeding
platelet/vessel
bleeding from superficial cuts
platelet disorder
delayed bleeding
coag disorder
hemarthroses
coag disorder
hematomas
coag disorder
PT measures
"extrinsic pathway, VII, warfarin"
Vit K is necessary for
"II, VII, IX, X, C and S"
PTT measures
"intrinsic path, VIII, IX, XI, XII, V, X, heparin"
Hemophilia A
"VIII disorder, long PTT"
Hemophilia B
"IX, long PTT"
Hemophilia C
"Auto recessive, XI disorder, long PTT"
PT is normal
"vWF, hemo A or B, thrombocytopenia"
PT increased
Vit K def
PTT normal or high
"vWF, vit K"
PTT is high
Heme A or B
Normal plate count
"vWF, hem A/B, Vit K def"
Low Plt
thrombocytopenia
What is the definition of cachexia
"Wasting syndrome resulting in altered metabolism, weight loss, loss of lean body mass, decreased albumin, increase CRP"
what area the clinical manifestations of cachexia
"nonvoluntary weight loss, anorexia, early satiety, hypermetabolism, loss of lean body mass/fat, anemia, weakness"
Progestagens
"inhibit cytokines, prevent NPY down regulation"
Thalidomide
blocks cytokine production
how does EPA counteract cachexia
"competes with arachadonic acid for COX and LOX, prevents synthesis of series 2 and 4 PG and TX. less inflamation less cachexia"
What physical exercise is recommended for cachexia pts
"resistance training 3 days/wk, 12 large muscle groups, 3 sets of 8"
Aflatoxin is linked to
HCC
Aromatic amines is linked to
Bladder cancer
Asbestos is linked to
Mesothelioma
Benzene is linked to
leukemia
Beryllium is linked to
Lung cancer
Vinyl Cloride is linked to
Angiosarcoma
What is a consequence of tumor cells having more laminin receptors
allow tumor cells to readily attack to organs
What is a major biochemical marker of cancer
"Keratin, IDs tumor as a carcinoma"
What are the signs of a neoplasm in a lymph node
"large painless, unilateral"
What factors are involved in the intrinsic path
"8,9,11,12"
PTT tests
intrinsic system by activating factor 12
What deficiency will PT/PTT not pick up
"factor 13, neither test looks at crosslinking of fibrin"
What factors is Vit K responsible for
"2, 7, 9, 10, S, C"
What will happen if bowel flora is suppressed
"less vit K synthesis, additive effect with coumadin"
What can you do to reverse a coumadin overdose
"wait it out, vit k, FFP, prothrombin factor"
What anticoag drug keeps a normal thrombin time
"coumadin.
In case of a major heparin overdose what is the Tx
protamine sulfate
DIC
"rapid coag with consumption of clotting factors resulting in thrombi, schistocytes, purpura. get secondary fibrinolysis"
What does DDimer measure
"fibrin degradation products. evidence that cross linking has occured. found elevated in DVT, PE"
How do you manage DIC
"Hydration, replacement therapy (for anything low ie plt), heparin, activated protein C (esp in cases of gram- infection)"
How does the liver relate to DIC
a failing liver will not remove excess clotting factors and a cascade can be set off more easily
DDVAP
"vasopressin analog, enhances vWF release, used for hemophilia and vW disease."
How is thrombocytopenia managed
"Corticosteroids, discontinue meds, IgG, IgRho, Plt transfusion"
How does Ig therapy fix thrombocytopenia
blocks removal of plates by the spleen
What are the 4 Ts of Heparin induced thrombocytopenia
"Thrombocytopenia, Timing (7-13 days post exposure), Thrombosis, Other causes of thrombocytopenia are not evident"
What occurs with TTP
"renal impairment, feer, neuro signs, hemolytic anemia, thrombocytopenia, "
What happens in TTP which does not happen in DIC
clumping of platelets and affecting kidney filtration. Lab tests are usually normal
What causes warfarin related skin necrosis
Protein C deficient Pt gets warfarin
What are the 4 major causes of anemia
"Marrow disorders, substrate deficiency, accelerated loss, decreased erythropoietin"
what is a normal Hb
"Men 14-16, women 12-15"
What is the normal ratio RBC:HgB:Hct
1:3:9
What is Hct
% of blood volume that is red cells
MCV
"how big cells are
"MCH, MCHC"
mean corpuscular hemoglobin and mean corpuscular hemoglobin concentration. heme per cell and heme per hct
RDW
"red cell distrobution width, 12-14"
WBC
5000-10000
Platelets
"150,000-450,000"
What changes in blood are seen in pregnancy
"RBC mass increase, RBC volume increase, but plasma increases more so may have low Hct"
what is a normal retic count
"25,000- 75,000"
when is a normal retic count not normal
"when the patient is anemic, retic count should be elevated."
What does a low retic count suggest
"bone marrow isnt working. Aplastic, suppressed, dysplastic"
what does a high retic count mean
marrow is working while pt is anemic (can be 6-7x higher)
What is the definition of a microcytic anemia
Less than 6 microns
What are the diseases with microcytic anemia
"Iron def
visually how can u call a cell hypochromic
if the central pallor is greater than 1/3 of the diameter of the cell
What are the 3 forms of iron in the body
"stored, useable, and transport"
As you begin to bleed where is iron lost first
plasma iron decreases briefly then iron is pulled from ferritin.
if bleeding continues and iron is depleted from storage what occurs
"TIBC goes up, and plasma ferritin goes down"
What gets fixed first during tx of iron deficient anemia
"serum iron, then transferrin, then ferritin."
B thalassemia trait
"usually asymptomatic, can have skeletal deformities due to massive marrow proliferation. MCV/RBC<13 suspicious. Codocytes (target cells)"
what happens biochemically in B thalassemia
"use up all the beta chains left with excess alpha chains, they glob together and precipitate out. inclusion bodies, splenomegaly"
Loss of 1 alpha trait
no complications
Loss of 2 alpha traits
still no symptoms can be cis or trans
lose 3 alpha genes
"hemoglobin H, splenectomy, transfusions"
Lose 4 alpha genes
"hydrops faetalis, not compatible with life"
Normocytic anemia
"Iron deficiency after a massive bleed, renal failure (less erythropoietin) marrow failure (slowed production)"
ACD
"disorder of iron homeostasis.
What are possible causes of ACD
"Arthritis, infections, cancer, lupus, bowel disease, sarcoid, vasculitis"
How do you stage renal failure
"based on glom filt rate.
What do you look for when you have pure RBC aplasia
Thyoma
Dacrocytes
"found in complete marrow aplasia, racket cells"
Fanconi's anemia
"aplastic, auto recess, defective DNA repair, cannot remove free radicals. fixed by marrow transplant"
Differential for macrocytic
"B12
Pernicious Anemia
"Anti intrinsic factor antibodies
hemolytic anemia
"immune regulated: Ab attack RBC. Coombs test. Looks for Ab either on the cells or in the blood.
what causes non immune regulated hemolytic anemia
"Artificial implants, heart valve abnormal, DIC, sickle cell, G6PD deficiency, "
Lab tests for hemolytic anemia
"bilirubin, indirect will be high. haptoglobin- will be low"
what are options in restoring iron
"ferrous sulfate, ferrous gluconate"
What can occur in iron overload
"GI pain, metabolic acidosis"
how do you treat iron overload
"gastric lavage, chelation using deferoxamine"
what happens in hemochromotosis
"intestines lose ability to stop absorbing iron, organ failure occurs"
why are cells macrocytic in folate deficiency
"get back up of FH3CH3, cells expand "
How are B12 and folate linked
cobalamin (B12) demethylates folate and is used to make methionine- used in myelin
what interacts with folate absorbtion
methotrexate and trimethoprim
Filgrastim
"G-CSF, increases production of neutrophils"
Mechlorethamine
"Alkylating, used in Lymphoma, ADR myelosuppression"
Mechlorethamine
"Alkylating, used in Lymphoma, ADR myelosuppression"
When do alkylating agents work
"anytime, cross link and alkylate the DNA (guanine mostly)"
When do alkylating agents work
"anytime, cross link and alkylate the DNA (guanine mostly)"
Cyclophosphamide
"alkylating, Burkitts, breast. Hemorrhagic Cystitis"
Cyclophosphamide
"alkylating, Burkitts, breast. Hemorrhagic Cystitis"
Ifosfamide
"alkylating, Burkitts, breast, Neurotoxicity"
Ifosfamide
"alkylating, Burkitts, breast, Neurotoxicity"
Chlorambucil
"Leukemia/Lymphoma, alkylating"
Chlorambucil
"Leukemia/Lymphoma, alkylating"
Busulfan
"Alkyl sulfonates, CML,
Busulfan
"Alkyl sulfonates, CML,
Carmustine
"Nitrosourea, HL, NHL, Brain, high lipid soluble, Renal tox"
Carmustine
"Nitrosourea, HL, NHL, Brain, high lipid soluble, Renal tox"
Cisplatin/Oxaliplatin
"Metal salt, G1, S phases. Damages renal tubes"
Cisplatin/Oxaliplatin
"Metal salt, G1, S phases. Damages renal tubes"
Doxorubicin
"Anthracyclines, Antibody, S/G2, Cardiac tox!!!!"
Doxorubicin
"Anthracyclines, Antibody, S/G2, Cardiac tox!!!!"
Bleomycin
"G2, HL, Testicular, lung, PULMONARY FIBROSIS"
Bleomycin
"G2, HL, Testicular, lung, PULMONARY FIBROSIS"
Methotrexate
"leads to polyglutination and cell death, S phase, need to alkalize urine"
Methotrexate
"leads to polyglutination and cell death, S phase, need to alkalize urine"
Mercaptopurine
S phase
Mercaptopurine
S phase
Cytarabine
"pyrimadine analog, S phase"
Cytarabine
"pyrimadine analog, S phase"
5 flurouracil
"S phase, bone marrow suppression"
5 flurouracil
"S phase, bone marrow suppression"
Vincristine/Vinblastine
"M phase, bind tubulin, cant go thru m phase. Neurotox"
Vincristine/Vinblastine
"M phase, bind tubulin, cant go thru m phase. Neurotox"
Etoposide
"Topoisomerase inhib. allows double strand breaks, S, G2 phase, 2ndary leukemia"
Etoposide
"Topoisomerase inhib. allows double strand breaks, S, G2 phase, 2ndary leukemia"
Paclitaxel
"polymerizes microtubules, M phase cant progress, G2/M. sever neurtopenia"
Paclitaxel
"polymerizes microtubules, M phase cant progress, G2/M. sever neurtopenia"
Asparaginase
"ALL, hypergycemia"
Asparaginase
"ALL, hypergycemia"
Tamoxifen
"inhibits estrogen receptors, no cell growth signalling"
Tamoxifen
"inhibits estrogen receptors, no cell growth signalling"
Leuprolide
"anti GnRH, stops LH, FSH release, used for prostate CA"
Leuprolide
"anti GnRH, stops LH, FSH release, used for prostate CA"
What cells are myeloid lineage
"Granulocytes, momocytes, platelets and erythrocytes"
what cells are lymphoid lineage
"B cells, T cells, NK cells"
What Il stimulates stem cells
1 and 6
What stimulates eosinophils
IL 5
In what cases do you worry about leukostasis
"AML WBC>100,000
what is a general blast marker
CD34
What disease is missing Factor VIII
Hemophilia A
What disease is missing factor IX
Hemophilia B
If you are vit K deficient what factors are affected
"2, 7, 9, 10"
What is occuring in acute leukemia
cells are not maturing completely
What defines AML
marrow is >20% blasts
what are the 2 major translocations in AML
t(15:17) and t(8:21)
what chemo agents typically cause a secondary AML
alkylating agents
what are factors in determining a prognosis in AML
"age, previous heme path, cytogenetics, performance status, MDR, high WBC at dx"
what is the typical chemo regimen for AML
7 and 3 anthracycline and cytarabine induction regimen.
what are the 3 stages of CLL
"chronic, accelerated, blast crisis"
What is Virchow's triad
"Coag State
what causes hemopoietic cells to become megakaryocytes
thrombopoietin
what produces TPO
liver and kidney
what is the receptor for TPO
CMPL
What is missing in Glanzmann thrombastenia
"Gp IIb/IIIa
What is missing bernard soulier syndrome
"GpIb
what sequence is important in plt binding
"RGD: arginine, glycine, aspartate"
alpha granule contains
"PF4, beta thromboglobulin, PDGF, vWF"
dense granules contain
ADP
what is gray platelet syndrome
no alpha granules
What do Omega 6 FA make
"Series 4 Leukotrienes
what are microparticles
tiny pieces of plts that are procoagulant. have encrypted tissue factor
"besides fibrinogen, what else can thrombin activate"
"V, VIII, XI, VII, XIII"
what does activated protein C do
inactivates V and VIII
what is a defining morphology of hodgkins
reed sternberg cells
what markers are on a stem cell in marrow
CD34
A genetic hit to a germinal center B cell gives rise to what forms of lymphoma
"follicular, LBL, HL, Burkitt"
Which bcl gene can lead to excessive cell production
BCL1
What bcl gene can lead to not enough apoptosis
BCL2
Changes in BCL1 are seen in what lymphoma
mantel cell lymphoma
Changes in BCL2 are seen in what lymphoma
follicular lymphoma
what is LDH used for
marker for cell turnover in lymphoma. lower levels show response to therapy
what does every lymphoma pt get
marrow biopsy
when is HL likely to occur
20-30s and 60-70s
what are B symptoms
"fever, night sweats, weight loss"
what factors attribute to a bad HL prognosis
"low albumin, low Hbg, male, stage IV, high WBC"
what is an important risk factor in NHL
Autoimmune disease and immunosuppression
SPEP is used for
finding imunoglobulins to determine subtype of lymphoma
what diseases are associated with NHL
"Sjorgrens, RA, SLE"
what is the prognostic index for follicular lymphoma and what goes into it
"FLIPI, number of sites, LDH, age over 60, Stage, low Hgb"
what is typical treatment for folicular lymphoma
watch and wait then rituximab
what does rituximab do
hones into CD20 and interferes w cell function
what is APLES
"the IPI for DLBCL, Age, performance, extranodal sites, LDH, stage"
What is the genetic translocation for M3 AML
t(15:17)
t(8:21) occurs in what leukemia
M2 AML
In what leukemia do you find auer rods
APML (M3)
what do you use to treat APML
"ATRA, except t(11:17)"
what markers are seen in ALL
"TdT, CD10, CD19"
what markers are seen in CLL
"CD20, 23, 5"
What stain is used in CML
LAP
"CD20, no CD5 means"
B cell prolymphocytic leukemia
TRAP stain is used to dx
Hairy Cell Leukemia
CD20 +"
Lymphocyte Predominant HL
CD10 BCL6
Burkitt's
CD3 with no 4 or 8
Sezary syndrome
Clover leaf and flower nuclei is a sign of
Adult T cell prolymphocytic leukemia
neutrophil infiltration into marginal zones
Nodal Marginal Zone Lymphoma
"lipid vacuoles, large macrophages, starry sky"
Burkitt's
"Lacunar RSC, collagenic fibrous bands"
Nodular Sclerosing HL
Popcorn cells (RSC)
Lymphocyte predominant HL
"Variation of follicle size, tingible Body macros"
Nonspecific follicular Hyperplasic
"asteroid bodies, schauman bodies, calcium oxalate crystals"
Sarcoid
"Macrophages with C shaped nuclei, monocytes with round nuclei"
Kikuchi
"Follicular hyperplasia, w granuloma of palisading macrophages"
Cat scratch or lymphogranuloma venerum
onionskin and lollipops
Castleman's
Foamy Macrophages
Whipples
Bland Macrophages
Virus associated Hemophagic syndrome
t(9:22)
"CML
7q21 deletion
Splenic marginal zone lymphoma
7q21 deletion
Splenic marginal zone lymphoma
t(14:18) bcl2 and cmyc
Follicular lymphoma
t(11:14) cyclin D
Mantle cell lymphoma
t(8:14)
Burkitt's
Trisomy 8
Hepatosplenic T cell lymphoma
What is used against Follicular lymphoma
R-CVP
what is used against LBCL
R CHOP
against HL what therapy agents are used
ABVD or the Stanford 5
what is at risk for and what is a richter transformation
"CLL, changes into LBCL"
Definition of myeloproliferative disorder
clonal expansion of a multipotent hematopoietic progenitor cell which leads to overproduction of one or more elements of blood
myelodysplasia
abnormal growth of myeloid line
How do you dx P vera
"exclusion, rule out hypoxia, renal disease, abnormal EPO receptor, if EPO level is low- look at PV"
what is erythromelagia and how is it treated
"erythema, pain, warmth, digital infarcts, NSAID"
What is myelofibrosis
"stem cell anomaly that leads to the overgrowth of one type of myeloid, in response there is an overgrowth of T cells and fibroblasts"
what is going to happen to marrow in AMM
hypervascular and fibrous scar tissue. start to see secondary sites of hematopoiesis
What are the hallmarks of AMM
"Marrow fibrosis, myeloid metaplasia, extramedularry hematopoiesis, 20% transform into AML"
What are the clinical features of AMM
"asymptomatic, splenomegaly, leukoerythroblastic smear, red bumpy skin"
What must be ruled to to dx as AMM
"CML, myelophthesis, leukemia, MPD"
Treatment for AMM
"treat sx, hydroxyurea, BMT, thalidomide"
Essential Thrombocytosis
"usually asymptomatic, jak+/-, rule out iron def, inflammation, CML (use LAP)"
How is ET treated
"anagrelide- poor tolerance
myelodysplastic syndromes
"ineffective erythropoiesis, like cytopenia, 33% becomes AML"
what are the qualities of a primary MDS
"idiopathic, simple genetic change, low IPSS, better tx response"
Secondary MDS have these qualities
"due to chemo, complex cytogenetics, High IPSS, poor outcomes"
RAEB-1
refractory anemia excess blasts. blasts <10%
RAEB-2
refractory anemia excess blasts. blasts >10%
RA
"reactive anemia >5% blasts and <15% sideroblasts, no auer rods"
RARS
>15% sideroblasts
RCMD
dysplasia in >10% abnormal in >1 cell lines
what mutation has a better outcome in MDS
"5q mutation, lenalidamide therapy"
Dx MDS
"Low blood count, macrocytic, r/o b12/folate def."
Dx MDA
"Low blood count, macrocytic, r/o b12/folate def."
tx MDS
"no standard. EPO, transfusion, GCSF, azacitidine"
What should a gamma spike look like in a healthy person
nice constant smooth slope
what does monoclonality mean
one type of Ig is over produced
what is linked to a worse outcome in MM
light chains in the urine
MGUS
"stable M spike, no light chains, no bone involvment, no renal faiure. can progress to MM"
Smoldering Myeloma
"M spike in between MM and MGUS, no bone, CA, or renal involvement. can transfrom to MM"
Solitary myeloma
"one tumor of plasma cells in the bone somewhere. no M spike, asymptomatic, Rad therapy"
Plasma Cell Leukemia
"russel and dutcher bodies, looks like CLL, hyperviscous blood, no skeletal, cold agglutination-raynaud's. very aggressive"
What are signs of MM
"low albumin, weak bones, kidney failure. CBC, BUN/creat, Ca, Ig level, UPEP, SKeletal survey, BM biopsy"
How do you treat stage II and III MM
"chemo, aggressive tx, BM transplant, bisphosphonates for bones (watch kidneys)"
What IL goes up in MM
IL 6
Components of Osteosclerotic Myeloma
"POEMS, demylenation, organomegaly, endocine, megakaryocytes, skin"