• 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

1/94

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;

94 Cards in this Set

  • Front
  • Back
What is the function of metabolism?
-elimination and detoxification of xenobiotics
-usually increase polarity
What are the classifications of metabolic reactions?
-phase I reactions-attach a small polar functional group (oxidations, reductions, and hydrolyses
-phase II reactions- attach a small polar molecule (glucuronidation, sulfation, glutathione conjugation, etc)
What are metabolic sites for drug metabolism?
-every tissue has some capability of drug metabolism
-most occurs in liver
-CYP's are in all cells of the body exceot red blood cels
What cells metabolize in the liver? kidney? lung? intestine? skin adn nose?
-hepatocytes (zonal distrbution)
-proximal tubular cells
-clara cells, type II cells
-mucosal cells and gut bacteria
-epithelial cells
What are the subcellular sites for drug metabolism?
-membrane-bound microsomes and mitochondria (endoplasmic reticulum, most pahse I enzymes located here)
-soluble- cytosol (most pahse II enzymes located here)
What are "expresed" enzymes? Preparation? advantages? disadvantages?
-enzyme is expressed in a cell line
-gene inserted into DNA of host cell
-grow cells and harvest
-isolate subcellular fractions
-can evaluate single isozyme activity, allows use of human enzymes, commercially available
-non-physiological, DNA sequence of gene must be known, technical challenges
Whata re isolated cells/.cultured cells? preparation? choice of technique? advantages? disadvanatages?
-good in vitro model
-measure metabolisma nd toxicity simultaneously
-organ is enzymatiically digested
-intact cells are collected and used immedatly or cultured
-freshly isolated cells or cultured cells (can differentiate and lose CYP enzymes)
-all metabolic pathways present, simultaneously measure metabolism and cytotoxicity, mutliple incubations from single animal, commercially available
-non-physiological, short lifespan (isolated cells), variable loss of enzyme acitivty (cultured cells)
What are subcellular fractions? preparation? advantages? disadvantages?
-measure metabolism in microsomes, cytosol, S-9 fraction or mitochondria
-prepare via differential centrifugation of organ homogenates
-separate ER from cytoplasm, can use any tissue in body
-easy preparation, simple meatbolic profile, multiple incubations froma single animal, commercially available
-non-physiological model, possible loss of activity with storage
Whata re purified enzymes? advanatges? disadvantages?
-purify from subcellular fractions, you cna find which CYP
-evaluate single isozyme activity, comemrcially available
-non-physiological, difficult purification
What are animal studies? advantages? disadvanatages?
-in vivo
-measure metabolites in tissues, plasma, urine, bile, feces, etc.
-all metabolic pathwasy are present, can measure pharmacokinetics (ADME)
-may require use of radiolabeled compound, expensive, biological variability, etc.
What is cytochrome p-450?
-enzyme found in virtually all eukaryotic and prokaryotic organisms (no subcellular organelles, cytoplasmic CYP's)
-several thousands of isozymes, mechanisms of action are same
-present in many tissues, found in all cells of body except RBC's
-microsomal, heme-containing enzyme (b/c CYP's are O2 binding, Hb in blood is not enzymatic
What is the general reaction of CYP 450? What does it require?
-commonly catalyses monooxygenations
-inserts single oxygen atom into substrate
-oxygen source- O2
-cofactor- B-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH)
-coenzyme-NADPH cytochrome p-450 reductase
-other- membrane lipids (help to keep the 2 enzymes close to each other
What are other reactions CYP 450 do?
-desaturations, reductions, etc.
Where is CYP isozyme 1A1/1A2 located? Typical substrates?
-many (1A1 low in liver
-polycyclic aromatic hydrocarbons (PAH)
-in cigarette smoke
Where is CYP isozyme 1B1 located? Typical substrates?
-many
-polycyclic aromatic hydrocarbons
Where is CYP isozyme 2A6 located? typical siubstrates?
-liver
-steroids
Where is CYP isozyme 2B6 located? typical substrates?
-liver, heart
-nicotine
Where is CYP isozyme 2C9/2C10 located? typical substrates?
-liver
-tolbutamide
Where is CYP isozyme 2C19 located? typical substrates?
-liver, heart
-omeprazole
Where is CYP isozyme 2D6 located? typical substrates?
-liver, heart, brain
-antidepressants and Beta-blockers
Where is CYP isozyme 2E1 located? Typical substrates?
-many
-acetaminophen (toxicity)
Where is CYP isozyme 2F located? typical substrates?
-lung
-coumarins
Where is CYP isozyme 3A4/3A5 located? typical substrates?
-many (adult)
-many
Where is CYP isozyme 3A7 located? typicla substrates?
-liver (fetal)
-many similar to 3A4
Where is CYP isozyme 4A9/4A11 located? typical substrates?
-kidney
-fatty acids
What percentage of CYP isozyme 1A2 exists? role in metabolism? inducers? inhibitors?
-13%
-<4%
-PAH's
-furafylline
What percentage of CYP isozyme 2A6 exists? role in metabolism? inducers? inhibitors?
-4%
-<1%
-phenobarbital
-no inhibitors
What percentage of CYP isozyme 2B6 exists? role in metabolism? inducers? inhibitors?
-1%
-<1%
-phenobarbital
-no inhibitors
What percentage of CYP isozyme 2C8, 2C9/2C10, and 2C19 exist? role in metabolism? inducers? inhibitors?
-20%
-11%
-phenobarbital
-tolbutamide (2C8), quercetin (2C8), sulfaphenazole (2C9/10), mephenytoin (2C19), and omeprazole (2C19)
What percentage of CYP isozyme 2D6 exists? role in metabolism? inducers? inhibitors?
-4%
-30%
-no inducers
-no inhibitors
What percentage of CYP isozyme 2E1 exists? role in metabolism? inducers? inhibitors?
-6%
-2%
-ethanol, isoniazid, and starvation
-diethldithiocarbamate
What percentage of CYP isozyme 3A4 exists? role in metabolism? inducers? inhibitors?
-30%
-52%
-rifampin
-troleandomycin and ketoconazole
What oxidative reactions are catalysed by cytochrome P-450?
-aliphatic hydroxylation
-aromatic hydroxylation
-epoxidation
-dealkylation
-oxidation on heteroatoms
What is aliphatic hydroxylation by cytochrome P450?
-occurs at C-H bonds in aliphatic chains (very stonrg bond)
What experiment is a good example of aliphatic hydroxylation by cytochrome P450 and what was significant about it?
-hydroxylation of valproic acid by rat liver microsomes
-animals treated with phenobarbital before incubated with microsomes
-had NADPH generating system (constanty generates NADPH so rxn isn;t limited)
-take out NADPH to see if metabolism still occurs, if it does, then it;s not CYP mediated
-carbon monoxide inhibited CYP even with phenobarbital induction
What is aromatic hydroxylation by cytochrome P450?
-occurs at C-H bond in aromatic ring
What experiment is a good example of aromatic hydroxylation by cytochrome P450 and what was significant about it?
-hydroxylation of warfarin by purified rat liver CYP isozymes
-incubation contained warfarin, CYP, CYP reductase, NADPH, and phospholipids
-isozyme 2C11 is the predominant isozyme in mlae rat liver, not found in female rat liver
-2C6 is induced by phenobarbital, also 1A1
-isozymes discriminate b/t metabolites produced, catalytic cylces are all identical though, probably depends on how warfarin orients itself on the active site of the enzyme
What is epoxidation by cytochrome P450?
-formation of epoxide ring at carbon-carbon double bonds and ona romatic rings
-monooxygenation
What experiment is a good example of epoxidation by cytochrome p450 and what was significant about it?
-epoxidation of acrylonitrile by isolated rat lung cells
-potentially carcinogenic to humans, can also be aersolized
-known carcinogen in rats
-cytochrome p450 has predminant role in making epoxide in specific cells, like the lungs
What is dealkylation by cytochrome p450?
-loss of an alkyl group
-monooxygenation, but doesn't look like one
-oxygen goes on alkyl group cleaved off and becomes an aldehyde
-necer occurs where carbon is surrounded by alkylic groups
What experiment is a good example of dealkylation by cytochrome p450? How is it significant?
-N-dealkylation of cisapride to nor cisapride
-CYP3A4 are having biggest effect therefore assume that CYP3A4 is the isozyme that metabolises cisapride
what is oxidation on heteroatoms by cytochrome P450?
-can occur on nitrogen or sulfur atoms
What is the experiment used as an example of oxidation on heteroatoms? How was it significant?
-N-oxidation of procainamide by human liver microsomes
-also done under N2 air and the presence of cimetidine
-also boiled to see if it's an enzymatic reaction, it is
-N2 not an inhibitor of enzyme, displaces O2 which makes the inhibitor not work, safer than CO
What are reductions done by cytochrome p450? Example?
-can occur when O2 tension is low
-may lead to formation of free radicals
-halothane dehalogenation (oxidation vs reduction)
-uses aliphatic hydroxylation, but 90% oxidative
-can cause liver damage
-What are desaturations by cytochrome p450? Example?
-direct conversion of single bond to double bond
-probably occurs via free radicals
-desaturation of valproic acid
What is flavin monooxygenase (FMO)?
-flavin containing enzyme
-microsomal, requires O2 and NADPH
-catalyses oxidations on nitrogens and sulfur atoms (never carbon)
-products may be identical to those produced by CYPs
What experiment is a good example of flavin monooxygenase? How is it significant?
-S-oxidation of thiobencard by striped bass liver microsomes
-thiourea was used as an FMO inhibitor and aminobenzotriazole was used as a ABT and CYP inhibitor
-FMO is sensisitve to heating
-metabolism wasn;t affectede by CYP inhibitor but by FMO inhibitor
What are molybdenum hydroxylases?
-include aldehyde oxidase (AO) and xanthine oxidase (XO)
-contain molybdenum, flavin, and iron in active site
-cytosolic, water is oxygen donor
-generally catalyse oxidations adjacent to a nitrogen or sulfur
-also CYP dependent
What experiment is a good example of molybdenum hydroxylases? How is it significant?
-oxidation of 6-oxypenciclovir by partially purified hepatic aldehyde oxidase
-menadione/isovanillin uses as aldehyde oxidase inhibitor and allopurinol as an xanthine oxidase inhibitor
-showed that xanthine oxidase is dependent on aldehyde oxidase
What is alcohol dehydrogenase (ADH)?
--distributed mostly in liver, also kidney and lung (cytosolic)
-cofactor is B-nicotinamide adenine dinucleoside (NAD+)
-oxidizes alcohols to aldehydes (reversible)
What is aldehyde dehydrogenase (AIDH)?
-distribution is mostly in liver
-cofactor is NAD+
-oxidizes aldehydes to carboxylic acids (irreversible)
What experiment is a good example of alcohol and aldehyde dehydrogenase? What is significant about it?
-pyrazole was used as a ADH inhibitor
-disulfiram was used as a AIDH inhibitor
-ADH is faster that ADIH
-so AIDH is dependent on ADH for reaction to complete
-so liver damage can occur if AIDH doesn't react
What is prostaglandin H synthase (PHS)?
-catalyses conversion of arachidonic acid to prostaglandin H2 (PGH2) via prostaglandin G2 (PGG2)
-extrahepatic (renal medulla)
-limited role in metabolism of xenobiotics
What experiment is a good example of prostanglandin H synthase? What is signifcant about it?
-renal metabolism of acetaminophen (APAP)
-indomethacin used as a PHS inhibitor
How is alcohol dehydrogenase a reductive enzyme? Example?
-catalyses reduction of aldehyde to alchohol
-suhc as reduction of chloral to trichloroethanol
What is mammalian aldo-keto reductase (liver)? example?
-reduction of a ketone to secondary alcohol
-reduction of naltrexone
What are bacterial reductases (intestinal)? Example?
-reduction of a azo group to an amino group
-reduction of sulfasalazine
What enzymes conduct hydrolyses?
-epoxide hydrolase (EH)
-esterases
-amidases
What is epoxide hydrolyase (EH)? Example?
-catalyses addition of water to epoxides forming dialcohols
-microsomal and cytosolic forms known
-widely distributed in the body (liver, kidney, lungs, intestines, etc)
-detoxifies potentially reactive epoxides
-hydrolyses of safrole epoxide
What are esterases? Example?
-catalyses the hydrolyses of esters to carboxylic acids and alcohols
-widely distributed in body (nicrosomes, cytosol, and blood plasma)
-occurs durign hydrolyses of aspriin to salicyclic acid
What are amidiases? Example?
-catalyse hydrolysis of amides to carboxylic acids and amines
-distribution somewhat more restricted than esterases
-occurs during hdyrolysis of procainamide to p-aminobenzoic acid
What are internal factors that affect drug metabolism?
-species/strain
-age
-gender
-hormonal affects
-genetic polymorphisms
How does the species/strain affect drug metabolism? What are the phase I enzymes for various species? Phase II enzymes?
-it can affect phase I or phase II pathways
-Phase I enzyme in humans in CYP3A4
-Phase I is male rats is CYP3A1/3A2
-Phase I in female rats is CYP3A9
-Phase II for cats is UGT1A6 deficiency and NAT1/NAT2 deficency in dogs
-phase II for pigs is SULT1A1
-strain-dependent differences most obvious in rodents
What specific experiments are examples of how species/strain affects drug metabolism? Significance?
-para vs ortho ainiline hydroxylation
-cats and dogs are ortho dominated
-ferrets are half and half
-mouse, rats, and gerbils are para dominant
-may be due to different shape in active sites of enzymes in different species
-hexobarbital-induced sleeping time in mice
-diiferent strains can metabolize hexobarbital faster than others
How does age affect drug metabolism?
-reduced biotransformation in fetal and newborn animals
-metabolism generally declines with age
-infants use CYP3A7 then CYP3A4 when grown up
What experiment is a good example of age affect drug metabolism? significance?
-effect of age on liver microsomes from female rats
-metabolism of certain enzymes slows age rats age
How does gender affect drug metabolism? What experiment is a good example? Significance?
-xenobiotic metabolism is generally similar in humans
-gender-related differences very common in rodents
-dealkylation of ethylmorphine
-metabolism differences can be seen in rats but not in other species
How do hormonal effects affect drug metabolism? Experimental examples? significances?
-usually very complex
-diabetes can be involves
-effect of streptozotocin (STZ)-induced diabetes on metabolism in male rats
-metabolism decreases even though CYP concentration stays the same
-other endocrine organs (adrenals, thyroid, etc.) can decrease metabolism if removes and can restore activity by repleacement therapy with appropriate hormones
-pregancy can be involved
-effect on metabolism in rats
-certain enzyme metabolism is decreased while other are increased
How do genetic polymorphisms affect drug metabolism?
-N-acetyltransferases (NATs) polymorphisms are well recognized
-different ethnic groups have a higher percentage of slower metabolism than other ethnicities
-cytochrome p450 is highly polymorphic
-glutathione S-transferase deficiency may be assoiciated with increased risk in certain cancers
-increased toxicity of irinotecan in UGT1A1-deficient individuals
How do environmental conditions affect drug metabolism? Experimental example? Significance?
-modification of natural circadian rhythms
-effect of light on CYP activity in rats
-increase sleep time leads to decreased metabolism and vice-versa
What other factors affect drug metabolism other than itnernal or environmental factors?
-enzyme inducers and inhibitors
-dietary diefiencies can have unpredictable effects
-such as: starvation (CYP2E1 induction), protein/fat, carbohydrates, vitamins, and minerals
-disease status of liver such as infections/tumors, and chronic ehtnaol abuse which decreases drug metabolism
What is glucuronidation?
-substrate is conjugated with glucuronic acid
-glucuronide conjugates are usually non-toxic
-common phase II pathway (depends on species)
-cofactor is readily available
-enzymes have wide substrate specificity
What is the mechanisms of the reaction of glucuronidation?
-involves a microsomal enzyme (very tightly membrane-bound) known as UDP-glucuronosyltranserfase (UGTs)
-isozymes are known (e.g. UGT1A1)
-reaction proceeds with inversion of stereochemistry
-cofactor is uridine-5'-diphospho-a-D-glucuronic acid (UDPGA)
-occurs on oxygen, nitrogen, sulfur (rare) and even carbon (very rare)
What specific reactions can occur during glucuronidation? Examples of each?
-O-glucuronidation can occur with alcohols, ohenols, and carboxylic acids (such as 1-naphthal)
-N-glucuronidation can occur with amines, amides, and sulfoamides (such as desipramine)
-S-glucuronidation (rare) can occur with thiol (-SH) (such as methimazole)
-C-glucuronidation (very rare) can occur on alkynes or 1,3 dicarbonyl groups (such as phenylbutazone)
What is sulfation?
-substrate is conjugated with sulfate ion (more polar)
-sulfate conjugates are usually non-toxic
-can compete with glucuronidation (depends on substrate and species
What is the general reaction for sulfation?
-requires cytosolic enzyme known as sulfotransferase (SULTs)
-isozymes are known (e.g SULT1E1-estrogen sulfotransferase)
-cofactor is 3'-phosphoadenosine-5'phosphosulfate (PAPs)
-such as acetaminophen
-occurs primarily with phenols and to a lesser extent with albohols
What is acetylation? What is the general reaction? Example?
-substrate is conjugated with an acetyl (COCH3) group (less polar)
-cytosolic enzymes known as N-acetyltransferases (NAT1 and NAT2)
-cofactor is acetyl coenzyme A (CH3CO-S-CoA)
-occurs most commonly with amines (esp. if attached to an aromatic ring)
-genetic polymorphism exists (slow vs. fast acetylators)
-such as acetylation of sulfamethazine
What is methylation? What is the general reaction? example?
-substrate is conjugated with a methyl group (CH3) (less polar)
-relatively rare for drugs, more common for endogenous substances
-involves enzyes nown as methyl transferases (cytosolic)
-cofactor is S-adenosylmethione (AdoMet or SAM)
-occurs on oxygen, nitrogen, or sulfur
-such as O-methylation of tea polyphenols by human placental catechol O-methyltransferase
What is amino acid conjugation? What is the general reaction? Example?
-substrate is conjugated with an amino acid (more polar)
-relatively minor pathway for most drugs
-species-dependent
-catalyzed dequentially by CoA synthetase and amino acid N-acryltransferase (mitochondrial)
-cofactors include ATP and coenzyme A
-occurs only with carboxylic acids
-such as glycine conjugation of salicyclic acid to salicycluric acid (70% total)
-amino acid conjugation of vaproic acid (VPA)
What is glutathione conjugation? General reaction? examples?
-substrate is conjugated with glutathione (less reaction and more polar)
-catalysed by glutathione S-transferase
-cytosolic and isozymes are known
-important detoxification pathway for epoxides and quinone
-also metabolizes acetaminophen for detoxification of NAPQI
What is interorgan metabolism of glutathione conjugates?
-conjugate probably formed in liver
-metabolized to cystein conjugate in bile duct/intenstines (y-glutamyltranspeptidase removes the glutamic acid and cysteinylglycine dipeptidase removes the glycine)
-cystein conjugate may be converted to an N-acetylcysteine conjugate (mercapturic acid)
-elimination or further metabolism may occur in the kidneys such as cystein and/or N-acetylcystein conjugates may be elimnated by urine or may be converted to a thiol conjugate for further metabolism
What is the anatomy/physiology of the liver?
-hepatocytes radiate out in "cords" from central veins
-extraction of solutes from sinusoidal blood
-secretion of drugs/metabolites from liver (blood vs bile)
-barrier of movement exists b/t blood and bile
-blood goes through sinusoidal membrane
-bile goes through canalicular membrane
What is the function of bile?
-digestion of lipids
-excretion of bilirubin(deggradation of hemeglobin) , excess cholesterol, etc.
What is the composition of bile?
-made up of water, ions, bile salts, GSH, phase II metabolites, etc.
-similar electrolyte composition to plasma, but essentially protein-free
What is the flow of bile?
-solute transport (biliary excretion of bromosulfophthalein) is actually active transport since it can become saturated
-osmotic movement of water through aqua porin channels
-concentrated/stored in gall bladder
-canalicular contractions are involved to push bile through channels
What is choleresis? Cholestasis?
-increase in bileflow
-decrease in bile flow
-can lead to drug-drug interactions (phenobarbital)
What are class A compounds secreted in bile?
-glucose, Na, K, Cl, Hg, Cs, Th, Co
What are class B compounds secreted in bile?
-bile salts, bilirubin, phase II conjugates, creatinine, oubain, Pb, As, Mn
What are class C compounds secreted in bile?
-sucrose, inulin, albumin, phosphates, phospholipids, Zn, Au, Fe, Cr
What factors can affect biliary secretion?
-polarity
-molecular weight threshold (species-dependent)
-such as effect on excretion of biphenyl/chorobiphenyls in rats
-higher molecular weights go to feces (from bile)
What hepatocyte transporters are in the basolateral membrane?
-sodium-taurocholate cotransporting polypeptide (solute carrier)
-organic anion transporting polypeptides (solute carrier)
-organic cation transporters (solute carrier)
-multidrug resistance-assoicated protein 3 (ATP binding cassette)
What hepatocyte transporters are in the canalicular membrane?
-bile salt export pump (solute carrier)
-p-glycoprotein (ATP bindng cassette)
-multidrug resistance-assoicated proteien 2 (ATP binding cassette)
What are biliary metabolites?
-undergo fecal excretion or intestinal reabsorption
-outcome depends on polarity and MW of compounds
-further metabolism is possible (such as GSH conjugates and glucuronides and sulfates)
-extent of enterohepatic recycling can alter half-life of drug in body (increased is enhanced toxicity while decreased reduces toxicitiy
-biliary excretion can be altered by choleresis, cholestasis, liver disease, and broad spectrum antibiotics
What techniques/examples are used with bile?
-bile duct cannulation (biliary and urinary excretion of desmethylnaproxen in rats)
-canalicular membrane vesicles (carrier-mediated uptake of quinoline antibiotics)